CA2878485A1 - 3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments - Google Patents

3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments Download PDF

Info

Publication number
CA2878485A1
CA2878485A1 CA2878485A CA2878485A CA2878485A1 CA 2878485 A1 CA2878485 A1 CA 2878485A1 CA 2878485 A CA2878485 A CA 2878485A CA 2878485 A CA2878485 A CA 2878485A CA 2878485 A1 CA2878485 A1 CA 2878485A1
Authority
CA
Canada
Prior art keywords
tetraen
estra
represent hydrogen
ethyl
methyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2878485A
Other languages
French (fr)
Inventor
Ulrich Bothe
Matthias BUSEMANN
Oliver Martin Fischer
Naomi BARAK
Andrea Rotgeri
Tobias Marquardt
Christian Stegmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Pharma AG
Original Assignee
Bayer Pharma AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48748225&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2878485(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bayer Pharma AG filed Critical Bayer Pharma AG
Publication of CA2878485A1 publication Critical patent/CA2878485A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J43/00Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • C07J43/003Normal steroids having a nitrogen-containing hetero ring spiro-condensed or not condensed with the cyclopenta(a)hydrophenanthrene skeleton not condensed
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/08Antiseborrheics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/28Antiandrogens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/32Antioestrogens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents

Abstract

The invention relates to AKR1C3 inhibitors and methods for the production thereof, the use thereof for treating and/or preventing diseases, and the use thereof for producing medicaments for treating and/or preventing diseases, in particular menstrual pain and endometriosis.

Description

3-SUBSTITUTED E.STRA-1,3,5(10),16-TETRAENE DERIVATIVES, METHODS FOR THE
PRODUCTION THEREOF, PHARMACEUTICAL PREPARATIONS CONTAINING SAME, AND
USE THEREOF FOR TI-IE PRODUCTION OF MEDICAMENTS
The invention relates to AKR1C3 inhibitors and to processes for their preparation, to their use for the treatment and/or prophylaxis of diseases and also to their use for preparing medicaments for the treatment and/or prophylaxis of diseases, in particular bleeding disorders and endometriosis.
AKR1C3 is a multifunctional enzyme and catalyses inter alio the reduction of 4-androstene-3,17-dione (a weak androgen) to testosterone (a potent androgen) and of oestrone (a weak oestrogen) to 1713-oestradiol (a strong oestrogen). In addition, the reduction of prostaglandin (PG) H2 to PGF2a and PGD2 to 9a,1113-PGF2 is inhibited (T. M. Penning et. al., 2006, 'Aldo-keto reductase (AKR) 1C3: Role in prostate disease and the development of specific inhibitors', Molecular and Cellular Endocrinology 248(1-2), 182 -191).
The local formation of oestradiol (E2) plays a central role in the initiation and progression of breast cancer disorders and endometriosis. Reduction of the tissue concentrations of oestrogens and in particular of oestradiol is achieved by therapeutic administration of aromatase inhibitors (to inhibit the formation of oestrogens from androgens) and of sulphatase inhibitors (to block the formation of oestrone from oestrone sulphate).
However, both therapeutic approaches have the disadvantage that systemic oestrogen concentrations are radically reduced (A. Oster et. al., J. Med. Chem. 2010, 53, 8176-8186).
Recently, it has been demonstrated experimentally that endometriotic lesions are capable of synthesizing oestradiol locally (B. Delvoux et al., J Clin Endocrinol Metab. 2009, 94, 876-883). For the subtype of ovarial endometriosis, an overexpression of AKR1C3 mRNA has been described (T.
Smuc et al., Mol Cell Endocrinol. 2009 Mar 25; 301(1-2): 59-64).
There is a great need to identify novel inhibitors of the enzyme aldo-keto reductase 1C3 (AICR1C3) (synonyms: type 5 1713-hydroxysteroid dehydrogenase or prostaglandin F
synthase), since inhibitors have potential for the treatment of hormone-dependent disorders such as, for example, endometriosis, but also for the treatment of hormone-independent disorders (M.C. Byrns, Y. Jin, T.M. Penning, Journal of Steroid Biochemistry and Molecular Biology (2010); A. L. Lovering et. al., Cancer Res 64(5), 1802-1810). In addition to endometriosis, this also includes prostate cancer (K. M. Fung et al., Endocr Relat Cancer 13(1), 169-180), prostate hyperplasia (R. 0. Roberts et al., Prostate 66(4), 392-404), endometrial carcinoma (T. L. Rizner et al., Mol Cell Endocrinol 2006 248(1-2), 126-135), polycystic ovary syndrome (K. Qin et al., J Endocrinol Metab 2006, 91(1), 270-276), lung carcinoma Q. Lan, et al., Carcinogenesis 2004, 25(11), 2177-2181), non-Hodgkin lymphoma = (Q. Lan et al., Hum Genet 2007, 121(2), 161-168), hair loss (L. Colombe et al., Exp Dermatol 2007, 16(9), 762-769), adiposity (P. A. Svensson et al., Cell Mol Biol Lett 2008, 13(4), 599-613), bladder carcinoma (J. D. Figueroa, Carcinogenesis 2008, 29(10), 1955-1962), chronic myeloid leukaemia (J. Birthwistle, Mutat Res 2009, 662(1-2), 67-74), renal cell carcinoma (J. T. Azzarello, Int J Clin Exp Pathol 2009, 3(2), 147-155), breast cancer (M. C. Byrns, J
Steroid Biochem Mol Biol 2010, 118(3), 177-187), premature sexual maturity (C.
He, Hum Genet 2010, 128(5), 515-527) and chronic obstructive pulmonary disease (S.
Pierrou, Am J
Respir Crit Care 2007, 175(6), 577-586).
Some inhibitors of AKR1C3 are known (review: Joanna M Day, Helena J Tutill, Atul Purohit and Michael J Reed, Endocrine-Related Cancer (2008) 15, 665-692 see also patents US20100190826 and W02007/100066). A steroidal substance that has been described is, for example, EM-1404, which is based on the oestratriene skeleton having a spirolactone unit in position 17 (F. Labrie et al. US Patent 6,541,463, 2003).
j__ _H 3 CH

H2N 411:1 Further steroidal AKR1C3 inhibitors having a lactone unit are found in P.
Bydal, Van Luu-The, F. Labrie, D. Poirier, European Journal of Medicinal Chemistry 2009, 44, 632-644.
Fluorinated oestratriene derivatives have been described in D. Deluca, G.
Moller, A. Rosinus, W. Elger, A. Hillisch, J. Adamski, Mol. Cell. Endocrinol. 2006, 248, 218 -224.
The compounds according to the invention are substances based on an oestra-1,3,5(10),16-tetraene skeleton substituted by an aromatic heterocycle in position 17. S. E.
Barrie et al.
US 5604213 describe 17-(3-pyridyl)estra-1,3,5(10),16-tetraen-3-ol, a related substance substituted at carbon atom 3 by a free hydroxyl group, only as a 17a-hydroxylase/C17-20 lyase (Cypl7A1) inhibitor.
=
.=========
= ,N
/

HO
17-(3-pyridyl)estra-1,3,5(10),16-tetraen-3-ol In addition, US 5604213 does not describe any 17-(3-pyridypestra-1,3,5(10),16-tetraene derivatives substituted by an alkoxy or an alkyl group in position 3. The substituents at the 3-position of the compounds according to the invention claimed in the present invention additionally comprise one or more functional groups such as, for example, carboxyl groups or hydroxyl groups, resulting in a further structural difference to the substances described in US
5604213. Surprisingly, it has now been found that the compounds according to the invention claimed herein are potent inhibitors of AKR1C3.
Estra-1,3,5(10),16-tetraene derivatives substituted by an aminocarbonyl (-CONH2) group in position 3 are described in US 2005/0203075 as antiproliferatively and antiangiogenically active, without reference to a concrete molecular target. However, these derivatives are not substituted by a heterocycle in position 17 of the estra-1,3,5(10),16-tetraene skeleton.
A review of 17-pyridyl- and 17-pyrimidylandrostane derivatives described as Cypl7A1 inhibitors is found in V. M. Moreira et al. Current Medicinal Chemistry, 2008 Vol. 15, No. 9.
Compounds according to the invention are the compounds of the formula (I) and their salts, solvates and solvates of the salts, and also the compounds comprised by (I) and mentioned hereinbelow as embodiments and their salts, solvates and solvates of the salts.
The present inventiOn provides compounds of the formula (I) AO*
R OW Hz (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyrimidin-4-yl, pyrazin-2-yl, pyrida7in-4-yl, pyridazin-3-yl, optionally mono- or disubstituted by fluorine, chlorine, nitrile, hydroxyl, carboxyl, trifluoromethyl, pentafluoroethyl, methoxy, ethoxy, thfluoromethoxy, -OCH2CF3, -SO2CH3, -S02CH2CH3, -(C=0)CH3, CI-Ca-alkyl, -CH2OH, -C(CH3)20H, -CONH2, -(C=0)NHCH3, -(C=0)NHCH2CH3, -(C=0)N(CF13)2, -SO2NH2, -SO2NHCH3, -SO2N(CH3)2.
RI represents -0-CRaRb -Y where Ra and Rb independently of one another represent represent hydrogen, methyl, ethyl, propyl, isopropyl, phenyl, 4-fluorophenyl, 3-fluorophenyl, 2-fluorophenyl, CH3-0-CH2-, -CH2CF3 or Ra and Rb together represent -(CH2)- where n = 2, 3, 4 or 5 or Ra and Rb together represent -CH2-0-CH2-, -CH2-NH-CH2-, -CH2-N(CH3)-CH2-, -CH2CH2-0-CH2CH2-, -CH2CH2-NH-CH2CH2-, -CH2CH2-N(CH3)-CH2CH2--0-CRcRd-CReRf-Y where Re, Rd, e K Rf represent hydrogen or Re, Rf represent hydrogen and RC, Rd independently of one another represent methyl, ethyl or together represent -(CH2)n- where n = 2, 3, 4, 5 or together represent -CH2-0-CH2- or -CH2CH2-0-CH2CH2- or RC, Rd represent hydrogen and Re, Rf independently of one another represent methyl, ethyl, CF3CH2- or together represent -(CH2)n- where n = 2, 3, 4, 5, =
-CHiCH2-0-CH2CH2-, -CH2CH2-1\111-CH2CH2-, -CH2CH2-N(CH3)-CH2CH2-, =
-CH2-0-CH2-= -CH2-NH-CH2-5 -CH2-N(CH3)-CH2- or Rd, Re, Rf represent hydrogen and RC represents methyl, ethyl, trifluoromethyl or Rc, Rd, Rf represent hydrogen and Re represents methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy or Rd, Rf represent hydrogen and Rc, Re independently of one another represent methyl, ethyl, trifluoromethyl, -0-CH2CH2CH2-Y, -0-CH2C(CH3)2CH2-Y, -0-CH2CH2C(CH3)2-Y, -0-CH2CH2CH(CH3)-Y, -0-CH2-CH(OH)-CH2-Y
-OCH2CH2CH2CH-Y, -CH2-Y, -CRgRh-CRIRLY where Rg' IV, RI represent hydrogen or Rg, Rh, IV represent hydrogen and Ri represents methyl, ethyl, trifluoromethyl or R-1 represent hydrogen and Rg, Rh represent methyl or together represent -CH2-, -CH2CH2-, -CH2CH2CH2- or Rg represents methyl and Rh, Ri represent hydrogen, -CH2CH2CH2-Y, -CH2CH2C(CH3)2-Y or -CH2CH2CH2CH2-Y and Y -CO2H, -OH, -(C=0)NH2, -(C=0)NHC1-4-alkyl, -S(=0)CH3 and their salts, solvates and solvates of the salts.
Preference is given to compounds of the formula (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyrimidin-4-yl, pyrazin-2-yl, pyridazin-4-yl, pyridazin-3-yl, optionally monosubstituted by fluorine, chlorine, nitrile, hydroxyl, carbOxyl, 'trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, -S02CH3, -(C=0)CH3, C i-C4-alkyl, RI represents -0-CRaRb ¨Y where Ra and Rb independently of one another represent represent hydrogen, methyl, ethyl, propyl, isopropyl, Phenyl, CH3-0-CH2-, CF3CH2-, -0-CRcRd-CReRf-Y where Re, Rd, e K Rf represent hydrogen or Re, Rd represent hydrogen and Re, Rf independently of one another represent methyl, ethyl, CF3CH2- or Rd, Re, Rf represent hydrogen and Re represent methyl, ethyl or Re, Rd, Rf represent hydrogen and Re represent methyl, ethyl, -0-CH2CH2CH2-Y, -0-CH2C(CH3)2CH2-Y, -0-CH2CH2C(CH3)2-Y, -0-CH2CH2CH(CH3)-Y, -0-CH2-CH(OH)-CH2-Y or -CRgRh-CWW-Y where Rg' Rh' W, RI represent hydrogen or Rg, Rh, W represent hydrogen and RI represents methyl, ethyl or IV represent hydrogen and Rg, Rh represent methyl or Rg represents methyl and Rh, le, Ri represent hydrogen and represents -CO2H, -OH, -(C=0)NH2, -(C=0)NHC1-4-alkyl and their salts, solvates and solvates of the salts.
Particular preference is given to compounds of the formula (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyridazin-4-yl, optionally monosubstituted by fluorine, carboxyl, trifluoromethyl, methyl RI represents -0-CR1le ¨Y where Ra abd Rb represent hydrogen or Ra represents hydrogen and Rb represents methyl, ethyl, phenyl or CH3-0-CH2--0-CReRd-CReRf-Y where Rd represent hydrogen and Re, Rf represent methyl -0-CH2CH2CH2-Y, -0-CH2C(CH3)2CH2-Y, -0-CH2-CH(OH)-CH2-Y or -CH2-CH2-Y and represents -CO2H, -OH, -(C=0)NH2, -(C=0)NHC1-4-alkyl and their salts, solvates and solvates of the salts.
Very particular preference is given to compounds of the formula (I) in which A represents 5-fluoropyridin-3-yl, 5-(trifluoromethyl)pyridin-3-yl, 5-carboxypyridin-3-yl, 6-methylpyridin-3-yl, pyrimidin-5-yl, 6-methylpyridazin-4-y1 and R1 represents -0-CRaRb ¨CO2H, -0-CR5Rb-(C=0)NH2, -0-CRaRb-(C=0)NHCH2CH3 where Ra and Rb represent hydrogen or Ra represents hydrogen and Rb represents methyl, ethyl, phenyl, CH3OCH2-, -0-CR'Rd-CReRf-CO2H where R', Rd represent hydrogen and Re, Rf represent methyl, -0-CH2CH2CH2-0H, -0-CH2CH2CH2-CO2H, -0-CH2C(CH3)2CH2-0H or -0-CH2-CH(OH)-CH2-0H
and their salts, solvates and solvates of the salts.
The invention furthermore provides the compounds 3-[17-(5-fhioropyridin-3-yDestra-1,3,5(10),16-tetraen-3-yl]propanoic acid ({1745-(trifluoromethyppyridin-3-yliestra-1,3,5(10),16-tetraen-3-ylloxy)acetic acid {[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy} acetic acid {[17-(6-methylpyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}acetic acid {[17-(pyrimidin-5-yl)estra-1,3,5(10),16-tetraen-3-ylioxylacetic acid 5-[3-(carboxymethoxy)estra-1,3,5(10),16-tetraen-17-yljnicotinic acid {[17-(6-methylpyridazin-4-yl)estra-1,3,5(10),16-tetraen-3-yl]oxyl acetic acid 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylioxy}-2,2-dimethylpropanoic acid 4-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxylbutanoic acid 2-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxylpropanoic acid 2- { [17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl] oxy butanoic acid 2- { [17-(5-fluoropyridin-3 -yl)estra-1,3,5(10),16-tetraen-3 -yl] oxy -3 -methoxypropanoic acid 2-{[17-(3-pyridyl)estra-1,3,5(10),16-tetraen-3-yl]oxylpropanoic acid 2- { [17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yli oxy propanami de 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propan-1-01 3- { [17-(5-fluoropyridin-3 -yl)e stra-1,3,5(10),16-tetraen-3 -yl] oxy -2,2-dimethylpropan-1-01 3- { [17-(5-fluoropyri din-3-yl)estra-1,3,5(10),16-tetraen-3-yl] oxy propan-1,2-diol { [17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}(phenyl)acetic acid N-ethyl-2-( {1745-(trifluoromethyl)pyridin-3-yl]estra-1,3,5(10),16-tetraen-3-ylloxy)acetamide and their salts, solvates and solvates of the salts.
It has been found that the estra-1,3,5(10),16-tetraene-3-carbonylamino derivatives provided by the invention act as AKR1C3 inhibitors. The compounds claimed show strong inhibition of AKR1C3 in vitro (IC50 values <200 nM) and in most cases even IC50 values < 50 nM.
Depending on their structure, the compounds according to the invention (cf., for example, Examples 10, 11, 12, 13, 14, 17 and 18) can exist in certain stereoisomeric forms (diastereomers). Accordingly the invention comprises the diastereomers and their respective mixtures. Pim such mixtures of diastereomers, the stereoisomerically uniform components can be isolated in a known manner.
If the compounds according to the invention can be present in tautomeric forms, the present invention comprises all tautomeric forms.
Preferred salts in the context of the present invention are physiologically acceptable salts of the compounds according to the invention. However, the invention also comprises salts which for their part are not suitable for pharmaceutical applications, but which can be used, for example, for the isolation or purification of the compounds according to the invention.
Physiologically acceptable salts of the compounds according to the invention include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, acetic acid, formic acid, trifluoroacetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Physiologically acceptable salts of the compounds according to the invention also include salts of customary bases, such as, by way of example and by way of preference, alkali metal salts (for example sodium salts and potassium salts), alkaline earth metal salts (for example calcium salts and magnesium salts) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms such as, by way of example and by way of preference, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, txiethanolamine, dicyclohexylamine, dimethylaminoethanol, procain, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine and N-methylpiperidine.
In the context of the invention, solvates are those forms of the compounds according to the invention which, in the solid or liquid state, form a complex by coordination with solvent molecules. Hydrates are a specific form of the solvates where the coordination is with water. In the context of the present invention, preferred solvates are hydrates.
Moreover, the present invention also comprises prodrugs of the compounds according to the invention. The term "prodrugs" includes compounds which for their part may be biologically active or inactive but which, during the time they spend in the body, are converted into compounds according to the invention (for example metabolically or hydrolytically).
In the context of the present invention, unless specified differently, the substituents have the following meanings:
C1-C4-Alkyl. represents a straight-chain or branched alkyl radical having 1 to 4 carbon atoms, by way of example and by way of preference methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, isobutyl.
The invention furthermore provides processes for preparing the compounds of the formula (I) according to the invention. The preparation of the compounds (I) according to the invention can be illustrated by the synthesis schemes below:
Some of the compounds according to the invention can be prepared as described in an exemplary manner for the synthesis of Example 1, starting with oestrone (Synthesis scheme 1):
The conversion of oestrone into Intermediate 1 is known from the literature (Tetrahedron Letters, 2003, 44, 3071 ¨ 3074 or Journal of Medicinal Chemistry, 1986, 29, 692 ¨ 698) or can be accomplished by reacting oestrone with 1,1,2,2,3,3,4,4,4-nonafluorobutane-1-sulphonyl fluoride in THF in the presence of potassium carbonate.
Intermediate 2 is prepared by reaction with ethyl acrylate using the Heck reaction (J. Org.
Chem., 1972, 37(14), 2320-2322). Preferably, the reaction is carried out using ethyl acrylate, triethylamine, tri-2-tolylphosphine and palladium(II) acetate in acetonitrile.
Intermediate 3 is synthesized by hydrogenation in the presence of palladium on carbon.
The conversion into Intermediate 4 is carried out using trifluoromethanesulphonic anhydride or N,N-bis(trifluoromethanesulphonypaniline in the presence of a base such as pyridine, 2,6-dimethylpyridine or 2,6-di-tert-butylpyridine or in the presence of a tertiary amine such as triethylamine or diisopropylethylamine or using alkali metal hexamethylsilazanes or lithium diisopropylamide (LDA) (J. Med. Chem., 1995, 38, 2463 ¨ 2471, J. Org. Chem., 1985, 50, 1990 ¨ 1992, J. Am. Chem. Soc., 2009, 131, 9014 ¨ 9019, Archiv der Pharmazie (Weinheim, Germany), 2001, 334, 12, 373 - 374). Preferred is the reaction with trifluoromethanesulphonic anhydride in the presence of 2,6-di-tert-butylpyridine in dichloromethane.
Intermediate 5 is prepared via the Suzuki reaction, which is known to the person skilled in the art. To this end, Intermediate 4 is reacted with 5-fluoropyridine-3-boronic acid or with a corresponding boronic ester such as, for example, a pinacol boronate, an MIDA
boronate (D.
M. Knapp et al. J. Am. Chem. Soc. 2009, 131, 6961) or with a trifluoroborate salt (G. A.
Molander et al., J. Org. Chem. 2009, 74, 973). Suitable for use as catalysts are a large number of palladium-containing catalysts such as, for example, tetrakis(triphenylphOsphine)palladium(0), bis(triphenylphosphine)palladium(H) dichloride or [1,3-bis(2,6-diisopropylphenyl)imidazol-2-ylidene1(3-chloropyridyppalladium(II) dichloride (CAS 905459-27-0). Alternatively, it is possible to employ a palladium-containing source such as, for example, palladium(II) acetate, palladium(II) chloride or Pd(dba)2 in combination with a phosphorus-containing ligand such as, for example, triphenylphosphine, SPhos (D. M.
Knapp et. al., J. Am. Chem. Soc. 2009, 131, 6961) or RuPhos (G. A. Molander, J. Org. Chem.
2009, 74, 973). Preference is given to the reaction with 5-fluoropyridine-3-boronic acid in the presence of bis(triphenylphosphine)palladium(II) chloride.
The preparation of Example 1 starting with Intermediate 5 takes place by hydrolysis of carboxylic esters, which is known to the person skilled in the art. Preference is given to the reaction with aqueous sodium hydroxide solution in THF and ethanol.

. = CH0 , CH, 0* A411 .
F /Ci W
O
F F
----.. F.,4 tr0 F
H F Intermediate 1 ---"A F
HO F F
\

CH
Olt H3C.0 H,c,.0 00 0_111 Fi 0 Intermediate 3 Intermediate 2 F
/ "N
,..., ..=

H3CN,0 .3c,0 0 Intermediate 4 o Intermediate 5 / "N

= H 00 0*
O
H
0 Example 1 Synthesis scheme 1 A further subset of the compounds according to the invention can be prepared as shown in an exemplary manner by the synthesis of Examples 2 to 7 (Synthesis scheme 2):
Starting with oestrone, Intermediate 6 is reacted by reaction with benzyl bromoacetate or benzyl chloroacetate in the' presence of a base such as, for example, sodium hydride, potassium *tert-butoxide, caesium carbonate or potassium carbonate in DMSO, 1-.
methylpyrrolidin-2-one, DMF or tetrahydrofuran. Preference is given to the reaction with benzyl bromoacetate in the presence of potassium carbonate in tetrahydrofuran.
Intermediate 7 is synthesized analogously to the preparation of Intermediate 4. Starting with Intermediate 7, the Example compounds 2 to 7 are then prepared using reaction conditions as in the preparation of Intermediate 5 by Suzuki reaction. Preference is given to using the appropriate aromatic nitrogenous boronic acids or pinacol boronates. Starting with Example 2,3,4,5,6 and 7, further example compounds can be prepared, as shown in an exemplary manner for the preparation of Example 19. To this end, Example compound 2 (where A is 5-(trifluoromethyppyridin-3-y1) is reacted with ethylamine in the course of an amide synthesis reaction, which is known to the person skilled in the art. Preference is given to using 1,1'-carbonyldiimidazole and imidazole hydrochloride in 2-methyltetrahydrofuran, as described in Organic Process Research & Development 2009, 13, 106-113.

CH0 0.
111*

y 0 Intermediate 6 HO
--SC) F

F

41*
1111...._S oyoSSH
HOro IOW 0 Intermediate 7 Examples 2,3,4,5,6,7 4.6111 Example 19 = Synthesis scheme 2 A further subset of the compounds according to the invention can be prepared as shown in an exemplary manner by the synthesis of Examples 8 to 18 (Synthesis scheme 3):
3 -Methoxyestra-1,3,5(10),16-tetraen-17-y1 trifluoromethanesulphonate (S.
Cacchi, E. Morera, Synthesis, 1986, 4, 320 ¨ 322) is prepared analogously to the preparation of Intermediate 5 by Suzuki reaction with the appropriate nitrogenous aromatic boronic acids or pinacol boronates.
Preference is given to using 5-fluoropyridine-3-boronic acid (for the preparation of Intermediate 8) or pyridine-3-boronic acid (for the preparation of Intermediate 10).
Intermediates 9 and 11 are prepared by reaction with boron tribromide in the presence of 2,6-lutidine in dichloromethane. Preparation of the example compounds takes place by reaction with the appropriately substituted alkyl halides in the presence of a base such as sodium hydride, potassium tert-butoxide, caesium carbonate or potassium carbonate. If alkyl chlorides are used, sodium iodide or potassium iodide may optionally be added.
Preferred is the reaction with potassium carbonate as base in the presence of potassium iodide or sodium iodide. If the radical R1 of the example compounds contains a carboxyl group, starting with Intermediates 9 and 11 alkyl halides substituted by a methyl carboxylate or ethyl carboxylate group are employed, and in a second step the esters are hydrolyzed by addition of aqueous sodium hydroxide solution.

--S

0-*
H

11,C
3 Intermediate 8,10 4.

0-*
HO
SS
Examples 8 to 18 Intermediate 9,11 In an unfoieseeable manner, the compounds according to the invention display a useful spectrum of pharmacological activity and advantageous pharmacokinetic properties. They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of diseases in humans and animals. For the purpose of the present invention, the term "treatment" includes prophylaxis. The pharmaceutical efficacy of the compounds according to the invention can be explained by its action as AKR1C3 inhibitor. Accordingly, the compounds according to the invention are particularly suitable for the treatment and/or prophylaxis of endometriosis, of uterine leiomyomas, of uterine bleeding disorders, of dysmenorrhoea, of prostate carcinoma, of prostate hyperplasia, of acne, of seborrhoea, of hair loss, of premature sexual maturity, of polycystic ovary syndrome, of breast cancer, of lung cancer, of endometrial carcinoma, of renal cell carcinoma, of bladder carcinoma, of non-Hodgkin lymphomas, of chronic obstructive pulmonary disease (COPD), of adiposity or of inflammatory pain.
The present invention furthermore provides the use of the compounds according to the invention for preparing a medicament for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
The present invention furthermore provides a method for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above, using an effective amount of the compounds according to the invention.
The present invention furthermore provides the use of the compounds according to the invention for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above.
The present invention furthermore provides the compounds according to the invention for use in a method for the treatment and/or prophylaxis of the disorders mentioned above.
The present invention furthermore provides medicaments comprising at least one compound according to the invention and at least one or more further active compounds, in particular for the treatment and/or prophylaxis of the disorders mentioned above. The following suitable active compounds for combinations may be mentioned by way of example and by way of preference: selective oestrogen receptor modulators (SERMs), oestrogen receptor (ER) antagonists, aromatase inhibitors, 17f3 HSD1 inhibitors, steroid sulphatase (STS) inhibitors, GnRH agonists and antagonists, kisspeptin receptor (KISSR) antagonists, selective androgen receptor modulators (SARMs), androgens, 5a-reductase inhibitors, selective progesterone receptor modulators (SPRMs), gestagens, antigestagens, oral contraceptives, inhibitors of mitogen activating protein (MAP) kinases and inhibitors of the MAP kinases kinases (Mkk3/6, Mek1/2, ' Erk1/2), inhibitors of the protein kinases B (PKBa/13/1;
Akt1/2/3), inhibitors of the phosphoinositide 3-kinases (PI3K), inhibitors of the cyclin-dependent kinase (CDK1/2), inhibitors of the hypoxia-induced signal path (HIF 1 alpha inhibitors, activators of prolyl hydroxylases), histone deacetylase (HDAC) inhibitors, prostaglandin F
receptor (FP) (PTGFR) antagonists and non-steroidal antiflammatory drugs (NSAIDs).
The compounds of the present invention may be combined, for example, with known anti-hyperproliferative, cytostatic or cytotoxic substances for the treatment of cancerous diseases.
In addition, the compounds according to the invention may also be employed in combination with radiotherapy and/or a surgical intervention.
As active compounds suitable for combinations, there may be mentioned by way of example:
131I-chTNT, abarelix, abiraterone, aclarubicin, aldesleukin, alemtuzumab, alitretinoin, altretamine, aminoglutethimide, amrubicin, amsacrine, anastrozole, arglabin, arsenic trioxide, asparaginase, azacitidine, basiliximab, BAY 80-6946, BAY 1000394, BAY 86-9766 (RDEA 119), belotecan, bendamustine, bevacizumab, bexarotene, bicalutamide, bisantrene, bleomycin, bortezomib, buserelin, busulfan, cabazitaxel, calcium folinate, calcium levofolinate, capecitabine, carboplatin, carmofur, carmustine, catumaxomab, celecoxib, celmoleukin, cetuximab, chlorambucil, chlormadinone, chlormethine, cisplatin, cladribine, clodronic acid, clofarabine, crisantaspase, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, darbepoetin alfa, dasatinib, daunorubicin, decitabine, degarelix, denileukin diftitox, denosumab, deslorelin, dibrospidium chloride, docetaxel, doxifluridine, doxorubicin, doxorubicin + oestrone, eculizumab, edrecolomab, elliptinium acetate, eltrombopag, endostatin, enocitabine, epirubicin, epitiostanol, epoetin alfa, epoetin beta, eptaplatin, eribulin, erlotinib, estradiol, estramustine, etoposide, everolimus, exemestane, fadrozole, filgrastim, fludarabine, fluorouracil, flutamide, formestane, fotemustine, fulvestrant, gallium nitrate, ganirelix, gefitinib, gemcitabine, gemtuzumab, glutoxim, goserelin, histamine dihydrochloride, histrelin, hydroxycarbamide, 1-125 seeds, ibandronic acid, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib, imiquimod, improsulfan, interferon alfa, interferon beta, interferon gamma, ipilimumab, irinotecan, ixabepilone, lanreotide, lapatinib, lenalidomide, lenograstim, lentinan, letrozole, leuprorelin, levamisole, lisuride, lobaplatin, lomustine, lonidamine, masoprocol, medroxyprogesterone, megestrol, melphalan, mepitiostane, mercaptopurine, methotrexate, methoxsalen, methyl aminolevulinate, methyltestosterone, mifamurtide, miltefosine, miriplatin, mitobronitol, mitoguazone, mitolactol, mitomycin, mitotane, mitoxantrone, nedaplatin, nelarabine, nilotinib, ralutamitle, nimotuzumab, nimustine, nitracrine, ofatumumab, omeprazole, oprelvekin, oxaliplatin, p53 gene therapy, paclitaxel, palifermin, palladium-103 seed, pamidronic acid, panitumumab, pazopanib, pegaspargase, PEG-epoetin beta (methoxy PEG-epoetin beta), pegfilgrastim, peginterferon alfa-2b, pemetrexed, pentazocine, pentostatin, peplomycin, perfosfamide, picibanil, pirarubicin, plerixafor, plicamycin, poliglusam, polyestradiol phosphate, polysaccharide-K, porfimer sodium, pralatrexate, prednimustine, procarbazine, quinagolide, radium-223 chloride, raloxifene, raltitrexed, ranimustine, razoxane, refametinib, regorafenib, risedronic acid, rituximab, romidepsin, romiplostim, sargramostim, sipuleucel-T, sizofiran, sobuzoxane, sodium glycididazole, sorafenib, streptozocin, sunitinib, talaporfin, tamibarotene, tamoxifen, tasonermin, teceleukin, tegafur, tegafur + gimeracil +
oteracil, temoporfin, temozolomide, temsirolimus, teniposide, testosterone, tetrofosmin, thalidomide, thiotepa, thymalfasin, tioguanine, tocilizumab, topotecan, toremifene, tositumomab, trabectedin, trastuzumab, treosulfan, tretinoin, trilostane, triptorelin, trofosfarnide, tryptophan, ubenimex, valrubicin, vandetanib, vapreotide, vemurafenib, vinblastine, vincristine, vindesine, vinflunine, vinorelbine, vorinostat, vorozole, yttrium-90 glass microspheres, zinostatin, zinostatin stimalamer, zoledronic acid, zorubicin.
Preferably, the present invention relates to medicaments comprising at least one compound according to the invention and one or more of the following active compounds, in particular for the treatment and/or prophylaxis of androgen receptor-dependent proliferative disorders:
LHRH (luteinizing hormone-releasing hormone) agonists, LHRH (luteinizing hormone-releasing hormone) antagonists, C(17,20)-lyase inhibitors, 5-a-reductase inhibitors type I, 5-a-reductase inhibitors type II, mixed 5-a-reductase inhibitors type I/II, a-radiation emitting radiopharmaceuticals for the treatment of bone metastases, such as radium-223 chloride, cyto statics, VEGF (vascular endothelial growth factor) kinase inhibitors, antigestagens, anti-oestrogens, EGF antibodies, ' oestrogens or other androgen receptor antagonists, poly (ADP-ribose) polymerase I inhibitors, or bispecific T-cell engagers (BiTE) coupled to a cell surface protein such as, for example, prostate-specific membrane antigen (PSMA).
The invention also relates to pharmaceutical preparations comprising at least one compound of the general formula I (or physiologically acceptable addition salts thereof with organic or inorganic acids) and the use of these compounds for preparing medicaments, in particular for the indications mentioned above.
The compounds can be used for the indications mentioned above, both after oral and after parenteral administration.
The compounds according to the invention can have systemic and/or local action. For this purpose, they can be administered in a suitable way, for example orally, parenterally, pulmonarly, nasally, sublingually, lingually, buccally, rectally, dermally, transdermally, conjunctivally, otically or as an implant or stent.
For these routes of application, the compounds according to the invention can be administered in suitable dosage forms.
Dosage forms that function according to the prior art, with rapid and/or modified release of the compounds according to the invention, containing the compounds according to the invention in crystalline and/or amorphisized and/or dissolved form, are suitable for oral administration, for example tablets (uncoated or coated tablets, for example with enteric coatings or coatings with delayed dissolution or insoluble coatings, which control the release of the compound according to the invention), tablets that disintegrate rapidly in the oral cavity or films/wafers, films/lyophylisates, capsules (for example hard-gelatin or soft-gelatin capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.
Parenteral administration can take place with avoidance of an absorption step (for example intravenously, intra-arterially, intracardially, intraspinally or intralumbally) or with inclusion of absorption (for example intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). Suitable dosage forms for parenteral administration are inter alia injection and infusion preparations in the form of solutions, suspensions, emulsions, lyophilisates or sterile powders.
Suitable dosage forms for the other routes of administration are, for example, pharmaceutical forms for inhalation (inter alia powder inhalers, nebulizers), nasal drops, solutions, and sprays; tablets for lingual, sublingual or buccal administration, films/wafers or capsules, suppositories, ear or eye preparations, vaginal capsules, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (such as, for example, patches), milk, pastes, foams, dusting powders, implants, intrauterine systems, vaginal rings or stents.
The compounds according to the invention can be converted into the stated dosage forms.
This can take place in a manner that is known per se, by mixing with inert, non-toxic, pharmaceutically suitable auxiliaries. These auxiliaries include inter alia vehicles (for example microcrystalline cellulose, lactose, mannitol), solvents (for example liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecylsulphate, polyoxysorbitan oleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (for example antioxidants such as, for example, ascorbic acid), colorants (for example inorganic pigments such as, for example, iron oxides) and taste and/or odour correctants.
The present invention further relates to medicinal products comprising at least one compound according to the invention, usually together with one or more inert, non-toxic, pharmaceutically suitable auxiliaries, and their use for the purposes stated above.
In the case of oral administration, the amount per day is from about 0.01 to 100 mg/kg of body weight. The amount of a compound of the general formula I to be administered varies over a wide range and can cover every effective amount Depending on the condition to be treated and the method of administration, the amount of the compound administered can be 0.01 - 100 mg/kg of body weight per day.
Nevertheless, it may optionally be necessary to deviate from the stated amounts, mainly depending on body weight, route of administration, individual response to the active substance, type of preparation and time point or interval in which administration takes place.
Thus, in some cases it may be sufficient to use less than the aforementioned minimum amount, whereas in other cases the stated upper limit must be exceeded. In the case of administration of relatively large amounts, it may be advisable to divide these into several individual doses throughout the day.

The percentdges in the following tests and examples are, unless stated otherwise, percentages by weight; parts are parts by weight. Proportions of solvents, dilution ratios and information about concentration for liquid/liquid solutions relate in each case to volume.
List of abbreviations, chemistry Abbreviations and acronyms:
DMF /V,N-dimethylformamide DMSO dimethyl sulphoxide hour(s) HPLC high-pressure, high-performance liquid chromatography LC-MS liquid chromatography-coupled mass spectroscopy ES-MS electrospray mass spectroscopy min minute(s) MS mass spectroscopy NMR nuclear magnetic resonance spectroscopy RT room temperature TFA trifluoroacetic acid THF tetrahydrofuran Purification of the compounds according to the invention In some cases the compounds according to the invention could be purified by preparative HPLC, for example using an autopurifier apparatus from Waters (detection of the compounds by UV detection and electrospray ionization) in combination with commercially available, prepacked HPLC columns (for example XBridge column (from Waters), C18, 5 1.1.m, 30 x 100 mm). The solvent system used was acetonitrile/water with addition of formic acid.
Further additives known to the person skilled in the art, such as, for example, ammonia, ammonium acetate or trifluoroacetic acid, may be used. Instead of acetonitrile, it is also possible to use, for example, methanol.
In some cases, the following method was used for preparative HPLC separation:

=
Systerh: Waters auto purification system: pump 2545, sample manager 2767, CFO, DAD 2996, ELSD 2424, SQD
Column: )(Bridge C18 5[1m 100x30 mm Solvent: A = H20 + 0.1% by volume formic acid (99%) B = Acetonitrile Gradient: 0-1 min 1% B, 1-8 mm 1-99% B, 8-10 min 99% B
Flow rate: 50 ml/min Temperature: RT
Injection: 1 x 2.5 ml Detection: DAD scan range 210-400 nm MS ESI+, ESI-, scan range 160-1000 m/z Freeze-drying or vacuum centrifugation was used for removing the HPLC solvent mixture. If the resulting compounds are present as TFA salts or formate salts, they can be converted by standard laboratory procedures known to the person skilled in the art into the respective free bases.
In some cases, the compounds according to the invention could be purified by chromatography on silica gel. For this purpose, for example, prepacked silica gel cartridges (for example from Separtis, Isolute Flash silica gel) in combination with the Flashmaster II
chromatograh (Argonaut/Biotage) and chromatography solvents or solvent mixtures such as, for example, hexane, ethyl acetate and dichloromethane and methanol were used.
Structural analysis of the compounds according to the invention:
In some cases, the compounds according to the invention were analysed by LC-MS:
In some cases, the following analytical method was used:
Instrument: Waters Acquity UPLC-MS SQD; column: Acquity UPLC BEH C18 1.7 50x2.1mm; mobile phase A: water + 0.1% by volume of formic acid (99%), mobile phase B:
acetonitrile; gradient: 0-1.6 min 1-99% B, 1.6-2.0 mm 99% B; flow rate 0.8 ml/min;
temperature: 60 C; injection: 2 [11; DAD scan: 210-400 nm The following symbols are used in the NMR data of the compounds according to the invention:

singlet doublet triplet quartet quin quintet multiplet br broad mc centred multiplet Synthesis of the compounds according to the invention:
Intermediate 1 17-0xoestra-1,3,5(10)-trien-3-y1-1,1,2,2,3,3,4,4,4-nonafluorobutane-1-sulphonate F
F
F F
F F
30.0 g (111 mmol) of oestrone and 46.0 g (222 mmol) of potassium carbonate were initially charged in 300 ml of THF. 40.2 g (133 mmol) of 1,1,2,2,3,3,4,4,4-nonafluorobutane-1-sulphonyl fluoride were added, and the mixture was stirred under reflux for 7 h and at RT for 18 h. Another 0.2 equivalents of 1,1,2,2,3,3,4,4,4-butane-l-sulphonyl fluoride were then added, and the mixture was heated under reflux for 4 h. The solid was filtered off and the filtrate was concentrated to half of its original volume, poured onto saturated sodium chloride solution and stirred for 35 min. The phases were then separated, the aqueous phase was extracted twice with ethyl acetate and the combined organic phases were washed with water and saturated sodium chloride solution and dried over sodium sulphate. The solvent was removed and the residue was triturated with hexane. This gave 63.6 g of a solid. C221-121F904S.
1H-NMR (300MHz, DMSO-d6): 8 [ppm]= 0.80 (s, 311), 1.27 - 1.64 (m, 6H), 1.66 -1.80 (m, 1H), 1.85 -2.12 (m, 3H), 2.18 - 2.43 (m, 3H), 2.81 -2.94 (m, 2H), 7.11 - 7.21 (m, 211), 7.43 (d, 1H).

Intermediate 2 =
(E)-Ethyl 3-(17-oxoestra-1,3,5(10)-trien-3-yl)acrylate A.6011 H3C 0 -r--1 A mixture of 500 mg (0.91 mmol) of 17-oxoestra-1,3,5(10)-trien-3-y1-1,1,2,2,3,3,4,4,4-nonafluorobutane-l-sulphonate, 181 mg of ethyl acrylate, 144 microlitres of triethylamine, 47 mg (0.15 mmol) of tri-2-tolylphosphine and 14 mg (0.06 mmol) of palladium(II) acetate in 8 ml of acetonitrile was heated in a microwave at 150 C / 100 watt. The mixture was combined with two analogous reaction batches (in each case starting with 1 g of 17-oxoestra-1,3,5(10)-trien-3-y1-1,1,2,2,3,3,4,4,4-nonafluorobutane-1-sulphonate). The combined mixture was filtered through Celite, poured into aqueous ammonium chloride solution and stirred for 30 min. The mixture was extracted three times with dichloromethane, washed with saturated sodium chloride solution, dried over sodium sulphate and concentrated.
Purification- by column chromatography on silica gel (hexane/ethyl acetate) gave 287 mg of a solid. C23112803.
1H-NMR (400MHz, DMSO-d6): [pprn]= 0.80 (s, 3H), 1.21 (t, 3H), 1.28 - 1.45 (m, 3H), 1.45 - 1.60 (m, 311), 1.69 - 1.80 (m, 111), 1.87 - 2.10 (m, 3H), 2.18 - 2.29 (m, 111), 2.32 - 2.45 (m, 2H), 2.79 - 2.88 (m, 211), 4.14 (q, 2H), 6.51 (d, 1H), 7.29 (d, 1H), 7.39 (s, 1H), 7.41 ¨ 7.45 (m, 1H), 7.54 (d, 1H).
Intermediate 3 Ethyl 3-(17-oxoestra-1,3,5(10)-trien-3-yl)propanoate Fi3C0 100 mg of Pd/carbon (10%) were added to 282 mg (0.80 mmol) of (E)-ethyl 3-(17-oxoestra-1,3,5(10)-trien-3-ypacrylate in 10 ml of ethyl acetate. At RT, hydrogen was introduced for 1 h. The cataryst was filtered off and the residue was washed with a total of 10 ml of ethyl acetate. Removal of the solvent under reduced pressure gave 278 mg (98% of theory) of product. C23H3003.
1H-NMR (300MHz, chloroform-d): [ppm]= 0.90 (s, 3H), 1.25 (t, 3H), 1.35 - 1.73 (m), 1.91 - 2.21 (m, 4H), 2.23 - 2.34 (m, 1H), 2.35 - 2.56 (m, 2H), 2.60 (t, 2H), 2.84 -2.97 (m, 4H), 4.14 (q, 2H), 6.95 (s, 1H), 7.00 (d, 1H), 7.22 (d, 1H).
Intermediate 4 Ethyl 3-(17-{1(trifluoromethyl)sulphony11 oxy} estra-1,3,5(10),16-tetraen-3-yl)prop an o ate OFF
CH3 `%0 F

0.25 ml (1.16 mmol) of 2,6-di-tert-butylpyridine was added dropwise to a solution of 274 mg (0.77 mmol) of ethyl 3-(17-oxoestra-1,3,5(10)-trien-3-yl)propanoate in 10 ml of dichloromethane. 0.16 ml (0.93 mmol) of trifluoromethanesulphonic anhydride was then added dropwise, and the mixture was stirred at RT for 24 h. The mixture was poured carefully into 70 ml of saturated sodium bicarbonate solution, the phases were separated, the aqueous phase was extracted twice with dichloromethane and the combined organic phases were washed with saturated sodium bicarbonate solution, twice with 1M aqueous hydrochloric acid solution, sodium chloride solution, dried over sodium sulphate and concentrated. This gave 522 mg of a brown oil. C24H29F305S. 1H-NMR (400MHz, chloroform-d, selected signals): 8 [ppm]= 1.00 (s, 3H). 1.25 (t, 3H), 1.80 (td, 1H), 1.85 - 1.98 (m, 2H), 2.04 -2.17 (m, 1H), 2.25 - 2.47 (m, 3H), 2.55 - 2.67 (m), 2.79 - 2.96 (m), 4.14 (q, 2H), 5.58 - 5.67 (m, 1H), 6.95 (s, 111), 7.00 (d, 1H), 7.19 (d, 1H).
Intermediate 5 Ethyl 3-(17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl)propanoate 522 mg (1.07 mmol) of ethyl 3-[17- [(trifluoromethyl)sulphonyl] oxy estra-1,3,5(10),16-tetraen-3-yl]propanoate were initially charged in 5 ml of toluene and 3 ml of ethanol. 212 mg (1.4 equivalents) of 5-fluoropyridine-3-boronic acid, 91 mg of LiC1, 1.3 ml of 2M aqueous sodium carbonate solution and 38 mg of bis(triphenylphosphine)palladium(II) chloride were then added, and the mixture was heated in a microwave at 120 C/300W for 90 min. The mixture was filtered, the phases were separated and the aqueous phase was extracted twcie with in each case 20 ml of ethyl acetate. The combined organic phases were washed with saturated sodium bicarbonate solution and saturated sodium chloride solution, dried over sodium sulphate and concentrated. Purification by column chromatography on silica gel (hexane/ethyl acetate) gave 108 mg of a yellowish oil. C28H32FN02. 1H-NMR
(300MHz, CHLOROFORM-d): 8 [ppm]= 1.04 (s, 3H), 1.25 (t), 1.2 - 1.90 (m), 1.91 - 2.04 (m, 1H), 2.08 - 2.26 (m, 2H), 2.26 - 2.51 (m, 3H), 2.61 (t, 2H), 2.82 - 2.97 (m), 4.14 (q, 2H), 6.07 - 6.12 (m, 1H), 6.91 - 7.05 (m, 2H), 7.22 (d, 1H), 7.40 (dt, 1H), 8.34 (d, 1H), 8.45 -8.51 (m, 1H).
Intermediate 6 Benzyl [(17-oxoestra-1,3,5(10)-trien-3-y1)oxyl acetate A mixture of 5.00 g (18 mmol) of oestrone, 3.5 ml of benzyl bromoacetate (22 mmol) and 7.67 g (55 mmol) of potassium carbonate and 70 ml of THF was heated under reflux overnight. Another 0.2 equivalent of benzyl bromoacetate was added, and the mixture was =
stirred under reflux for a further 3 h. Water was added to the reaction mixture. The phases were allowed to separated, and the aqueous phase was extracted twice with ethyl acetate. The combined org. phases were washed with water, saturated sodium chloride solution, dried over sodium sulphate, filtered and concentrated. Purification by column chromatography on silica gel (hexane/ethyl acetate) gave 7.91 g of a colourless oil. C27H3004. 1H-NMR
(300MHz, chloroform-d): 8 [ppm]-- 0.91 (s, 311), 1.35 - 1.73 (m), 1.90 - 2.59 (m), 2.78 - 2.96 (m, 2H), 4.64 (s, 211), 5.24 (s, 2H), 6.60¨ 6.67 (m, 1H), 6.70 (dd, 111), 7.19 (d, 111), 7.35 (s, 5H).
Intermediate 7 Benzyl [(17- ff(triflu orom ethyl)sulp honyl] oxy} estra-1,3,5(10),16-tetraen-3-y 1)oxy] acetate 0 j --S

Oyc) O. H-7.9 g (18.9 mmol) of benzyl [(17-oxoestra-1,3,5(10)-trien-3-yDoxy]acetate were reacted analogously to the preparation of Intermediate 4 to give 10.1 g of the title compound as a crude product. C28H29F306S. 1H-NMR (400MHz, DMSO-d6): 8 [ppm].= 0.92 (s, 3H), 1.29 -1.61 (m), 1.65 - 1.86 (m, 3H), 2.02 - 2.15 (m, 111), 2.17 - 2.40 (m, 311), 2.66 - 2.78 (m, 2H), 4.76 (s, 2H), 5.15 (s, 2H), 5.72 (d, 1H), 6.57 (d, 111), 6.65 (dd, 1H), 7.10 (d, 1H), 7.27 - 7.41 (m, 51-1).
Intermediate 8 17-(5-Fluoropyridin-3-y1)-3-methoxyestra-1,3,5(10),16-tetraene N F

H3c, 410 1.23 g (0.05'equiv.) of bis(triphenylphosphine)palladium(II) chloride were added to a mixture of 14.6 g (35.1 mmol) of 3-methoxyestra-1,3,5 (10),16-tetraen-17-yltrifluoromethanesulpho-nate (S. Cacchi, E. Morera, Synthesis, 1986, 4, 320 ¨ 322), 6.92 g (1.4 equiv.) of 5-fluoropyridine-3-boronic acid, 2.97 g (2.0 equiv.) of lithium chloride, 47 ml of 2M aqueous sodium carbonate solution, 80 ml of ethanol and 100 ml of toluene, and the mixture was heated under reflux for 4.5 h. The mixture was filtered through Celite, the phases were separated and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with saturated aqueous sodium bicarbonate solution and sodium chloride solution, dried over sodium sulphate and concentrated. The residue was purified by column chromatography on silica gel (hexane/ethyl acetate). This gave 10.4 g (81% of theory) of a solid. C24H26FN0. 1H-NMR (300M'Hz, chloroform-d): 6 [ppm]= 1.05 (s, 3H), 1.40 -1.75 (m), 1.82 (td, 1H), 1.90 - 2.03 (m, 1H), 2.03 - 2.27 (m), 2.27 - 2.57 (m), 2.84 -3.03 (m, 2H), 3.79 (s, 3H), 5.99 - 6.18 (m, 1H), 6.66 (d, 1H), 6.73 (dd, 1H), 7.21 (d, 1H), 7.40 (dt, 1H), 8.34 (d, 1H), 8.48 (s, 1H).
Intermediate 9 17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol N

In a temperature range of from 0 C to 3 C, 11.6 ml of 2,6-lutidine diluted with 50 ml of dichloromethane were added dropwise to 100 ml of a 1M solution of boron tribromide in dichloromethane. 10.4 g of 17-(5 -fluoropyridin-3 -y1)-3 -methoxyestra-1,3 ,5 (10),16-tetraene dissolved in 50 ml of dichloromethane were then added dropwise at from 0 C to 3 C, and the mixture was allowed to warm to RT in an ice bath overnight. The reaction mixture was added to 400 ml of ice/water and stirred for 40 min. The phases were separated, the aqueous phases were extracted three times with in each case 50 ml of dichloromethane and the combined org.
phases were washed with saturated sodium chloride solution, dried over sodium sulphate and concentrated. The crude product was triturated at 40 C in hexane, filtered off with suction and =
washed with hexane. This gave 13.9 g of a brown solid (crude product).
C23H24FN0. MS
(ESIpos) mass found: 349.00.
1H-NMR (300MHz, DMSO-d6): 6 [ppm]= 0.98 (s, 3H), 1.27 - 1.61 (m, 4H), 1.69 (td, 1H), 1.78 ¨ 1.90 (m, 1H), 2.03 - 2.35 (m, 5H), 2.65 - 2.84 (m, 2H), 6.23 ¨6.28 (m, 1H), 6.40 - 6.52 (m, 2H), 7.01 (d, 1H), 7.63 - 7.70 (m, 1H), 8.42 (d, 1H), 8.46 ¨ 8.50 (m, 1H), 8.98 (s, 1H).
Intermediate 10 3-Methoxy-17-(3-pyridyl)estra-1,3,5(10),16-tetraene N

O

At 80 C, 3.00 g (7.20 mmol) of 3 -methoxyestra-1,3 ,5 (10),16-tetraen-17-yltrifluoromethanesulphonate were reacted analogously to Example 8 with 1.24 g (1.40 equiv.) of pyridine-3-boronic acid overnight analogously. This gave, after purification by column chromatography on silica gel (hexane/ethyl acetate), 1.2 g of the title compound as a crude product. 120 mg of the crude product were purified further by HPLC. This gave 60 mg of the title compound. C24H27N0. MS (ESIpos) mass found: 345.21. 1H-NMR
(400MHz, DMSO-d6): 6 [ppm]= 0.98 (s, 3H), 1.32 - 1.61 (m, 4H), 1.71 (td, 1H), 1.83 -1.91 (m, 111), 2.04 - 2.14 (m, 2H), 2.17 - 2.38 (m, 3H), 2.75 - 2.89 (m, 2H), 3.66 (s, 3H), 6.11 (dd, 1H), 6.60 (d, 1H), 6.65 (dd, 1H), 7.13 (d, 1H), 7.29 - 7.34 (m, 1H), 7.76 (dt, 1H), 8.42 (dd, 1H), 8.59 (d, 1H).
Intermediate 11 17-(3-Pyridyl)estra-1,3,5(10),16-tetraen-3-ol N

0-*

HO
Analogously to the preparation of Intermediate 9, 1.1 g of 3-methoxy-17-(3-pyridyl)estra-1,3,5(10),16-tetraene were reacted with boron tribromide (1M in dichloromethane) and 2,6-lutidine. For work-up, the mixture was poured into into ice-water and stirred.
Dichloromethane was added. The solid that remained was filtered off with suction, washed with water and dichloroethane and dried by adding toluene followed by removal of the toluene on a rotary evaporator (three times). Trituration with diethyl ether and drying under reduced pressure gave 511 mg of the title compound. C23H25N0. MS (ESIpos) mass found:
332.00. 1H-NMR (400MHz, DMSO-d6): 8 [ppm]--- 1.01 (s, 3H), 1.31 - 1.61 (m, 4H), 1.72 (td, 1H), 1.85 (dt, 1H), 2.09 - 2.23 (m, 3H), 2.27 - 2.41 (m, 2H), 2.66 - 2.83 (m, 2H), 6.40 ¨ 6.45 (m, 2H), 6.49 (dd, 1H), 7.02 (d, 1H), 7.84 (dd, 1H), 8.37 ¨ 8.42 (m, 1H), 8.67 (d, 1H), 8.82 (s, 1H).
Preparation of the example compounds Example 1 3-117-(5-Fluoropyridin-3-ypestra-1,3,5(10),16-tetraen-3-yl]propanoic acid HO

A mixture of 100 mg (0.23 mmol) of ethyl 3-[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]propanoate, 3 ml of THF, 0.5 ml of ethanol and 0.58 ml of 2M
aqueous sodium hydroxide solution was stirred at RT overnight and then diluted with water and adjusted to a =
pH of 3 using a 10% strength aqueous citric acid solution. The aqueous phase was extracted three times with ethyl acetate. The combined organic phases were dried over sodium sulphate and concentrated. The residue was purified by preparative HPLC
(acetonitrile/water/formic acid). This gave 44 mg of a white solid. C26H28FN02. MS (ESIpos) mass found:
405.21. 111-NMR (300MHz, DMSO-d6): 8 [ppm]= 0.99 (s, 3H), 1.24 - 1.62 (m, 4H), 1.62 - 1.78 (m, 1H), 1.78 - 1.96 (m, 111), 2.01 - 2.40 (m, 5H), 2.70 (t, 2H), 2.75 - 2.93 (m, 2H), 6.26 (s., 1H), 6.85 -6.96 (m, 2H), 7.13 (d, 1H), 7.68 (d, 1H), 8.43 (d, 1H), 8.48 (s, 1H)., 12.1 (s).
Example 2 ({17- [5-(T rifluorom ethyl)pyridin-3-yl] estra-1,3,5(10),16-tetra en-3-y1) oxy)acetic acid FlO)ro 0 200 mg (0.36 mmol) benzyl { [17- { [(trifluoromethypsulphonyl]oxy} estra-1,3,5(10),16-tetraen-3-yl]oxy} acetate were initially charged in 1.5 ml of toluene and 1 ml of ethanol. 97 mg (1.4 equivalents) of 5-(trifluoromethyl)pyridine-3-boronic acid, 31 mg of LiC1, 0.49 ml of 2M aqueous sodium carbonate solution and 12 mg of 1,3-bis(2,6-diisopropylphenyl)imidazol-2-ylidene(3-chloropyridyl)palladium(II) dichloride (CAS 905459-27-0) were then added, and the mixture was heated in a microwave at 120 C/300W for 90 min. The mixture was filtered, 0.90 ml of 2 M aqueous sodium hydroxide solution was added and the mixture was heated in a microwave at 120 C/300W for 30 min. The reaction mixture was adjusted to a pH of 3 using 10% strength aqueous citric acid solution and the aqueous phase was extracted three times with ethyl acetate. The combined organic phases were washed with saturated sodium chloride solution, dried over sodium sulphate and concentrated. Purification by preparative HPLC
(acetontrile/water/formic acid) gave 43 mg of a solid. C26H26F3NO3. MS
(ESIpos) mass found: 457.19. 1H-NMR (300MHz, DMSO-d6): ö [ppm]= 1.00 (s, 3H), 1.27¨ 2.41 (m), 2.70 ¨ 2.88 (m, 2H), 4.56 (s, 2H), 6.35 (s., 1H), 6.46 - 6.73 (m, 2H), 7.13 (d, 1H), 8.03 (s., 1H), 8.83 (s, 1H), 8.90 (s, 1H), 12.9 (s).

=
Example 3 ' ([17-(5-Fluoropyridin-3-ypestra-1,3,5(10),16-tetraen-3-yl] oxy} acetic acid HO.ro Analogously to the preparation of Example 2, 200 mg (0.36 mmol) of benzyl {[17-{ [(trifluoromethypsulphonyl]oxyl estra-1,3,5(10),16-tetraen-3-yl]oxyl acetate were reacted with 72 mg (0.51 mmol) of 5-fluoropyridine-3-boronic acid. This gave 31 mg (21% of theory) of a solid. C25H26FN03. MS (ESIpos) mass found: 407.19. 1H-NMR (300MHz, DMSO-d6):
[ppin]= 0.98 (s, 3H), 1.26 - 1.63 (m, 4H), 1.64 ¨ 1.78 (m, 1H), 1.79 ¨ 1.93 (m, 1H), 2.00 -2.39 (m, 5H), 2.73 - 2.89 (m, 2H), 4.56 (s, 2H), 6.26 (s., 1H), 6.53 - 6.66 (m, 2H), 7.13 (d, 1H), 7.68 (dd, 1H), 8.42 (d, 1H), 8.48 (s, 1H), 12.9 (s).
Example 4 1[17-(6-Methylpyridin-3-yOestra-1,3,5(10),16-tetraen-3-yll oxy} acetic acid HO S. -1:1 9.6 mg of bis(triphenylphosphine)palladium(II) chloride were added to a mixture of 150 mg of benzyl [(17- { [(trifluoromethypsulphonyl] oxy estra-1,3,5(10),16-tetraen-3-yDoxy]ae,etate, 52 mg (1.4 equiv.) of 2-methyl-5-boronic acid, 23 mg (2.0 equiv.) of lithium chloride in 1.5 ml of toluene, 1 ml of ethanol and 0.37 ml 2M aqueous sodium carbonate solution, and the =
mixture was stirred at 100 C overnight. 0.7 ml of 2M aqueous sodium hydroxide solution was added, and the mixture was stirred at 100 C overnight. The reaction mixture was diluted with water, adjusted to pH = 4 with citric acid solution and extracted three times with ethyl acetate.
The combined organic phases were concentrated and purified by preparative HPLC. This gave 24 mg of the title compound. C26H29NO3. MS (ESIpos) mass found: 403.21. III-NMR
(300MHz, DMSO-d6): 8 [ppm]--- 0.96 (s, 3H), 1.29 - 1.62 (m, 411), 1.69 (td, 1H), 1.80 ¨ 1.92 (m, 111), 1.99 - 2.39 (m, 6H), 2.41 (s, 3H), 2.71 - 2.89 (m, 211), 4.56 (s, 2H), 6.00 ¨ 6.07 (m, 1H), 6.56 (d, 1H), 6.59 - 6.66 (m, 111), 7.10 - 7.22 (m, 2H), 7.61 - 7.68 (m, 1H), 8.44 (d, 114), 12.9 (hr. s, 1H).
Example 5 {117-(Pyrimidin-5-yl)estra-1,3,5(10),16-tetraen-3-ylloxy}acetic acid N

F10,1r0 01 Analogously to Example 4, 150 mg of benzyl [(17-{[(trifluoromethypsulphonyl]oxylestra-1,3,5(10),16-tetraen-3-ypoxy]acetate were reacted with 47 mg of pyrimidine-5-boronic acid.
Purification by preparative HPLC gave 42 mg of the title compound. C24H26N203.
MS
(ESIpos) mass found: 390.19. 111-NMR (400MHz, DMSO-d6): ö [ppm1= 0.98 (s, 3H), 1.33 -1.62 (m, 4H), 1.72 (td, 1H), 1.83 - 1.91 (m, 1H), 2.08 - 2.16 (m, 2H), 2.18 -2.38 (m, 3H), 2.62 - 2.65 (m, 1H), 2.73 - 2.87 (m, 2H), 4.55 (s, 2H), 6.27 - 6.31 (m, 1H), 6.57 (d, 1H), 6.63 (dd, 1H), 7.13 (d, 111), 8.82 (s, 211), 9.04 (s, 1H).
Example 6 5[3-(Carboxymethoxy)estra-1,3,5(10),16-tetraen-17-yllnicotinic acid =

HO

FlOo Analogously to Example 4, 150 mg of benzyl [(17-{[(trifluoromethypsulphonyl]oxylestra-1,3,5(10),16-tetraen-3-ypoxylacetate were reacted with 100 mg of methyl 544,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)nicotinate. Purification by preparative HPLC and trituration of the solid obtained with diethyl ether gave 39 mg of the title compound.
C26H27I\105. MS (ESIpos) mass found: 433.19. 1H-NMR (300MHz, DMSO-d6): [ppmJ=
0.99 (s, 3H), 1.30 - 1.63 (m, 5H), 1.63 - 1.79 (m, 1H), 1.82 - 1.93 (m, 1H), 2.04 -2.40 (m, 6H), 2.73 - 2.85 (m, 2H), 4.56 (s, 2H), 6.23 ¨ 6.27 (m, 1H), 6.57 (d, 1H), 6.63 (dd, 1H), 7.13 (d, 1H), 8.16 (t, 1H), 8.81 (d, 1H), 8.91 (d, 1H), 13.2 (br. s).
Example 7 ([17-(6-Methylpyridazin-4-yl)estra-1,3,5(10),16-tetraen-3-ylloxy}acetic acid HC

Fi0). SO H
(0 Analogously to Example 4, 181 mg (0.33 mmol) of benzyl [(17-{ [(trifluoromethypsulphonyl] oxy} estra-1,3,5 (10),16-tetraen-3 -yl)oxy]
acetate were reacted with 101 mg (0.46 mmol) of 3-methy1-5-(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-yl)pyridazine. Purification by preparative HPLC gave a crude product which was triturated with diethyl ether and with ethyl acetate. This gave 14 mg (9% of theory) of the title =
compound. C25H28I\103. MS (ESIpos) mass found: 404.21. 1H-NMR (400MHz, DMSO-d6):
[ppm]-= 1.01 (s, 3H), 1.32 - 1.61 (m, 4H), 1.69 (td, 1H), 1.82 - 1.90 (m, 1H), 2.08 - 2.38 (m, 5H), 2.58 (s, 3H), 2.71 - 2.88 (m, 2H), 4.51 (s, 2H), 6.54 - 6.58 (m, 2H), 6.62 (dd, 1H), 7.13 (d, 1H), 7.48 (d, 1H), 9.10 (d, 1H).
Example 8 3-1[17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylloxy}-2,2-dimethylpropanoic acid N F

H0)1)C0 A mixture of 120 mg (0.31 mmol) of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 61 mg (0.37 mmol, 1.2 equiv.) of methyl 3-ehloro-2,2-dimethylpropanoate, 212 mg (1.53 mmol, 5.0 equiv.) of potassium carbonate and 5 mg of potassium iodide in 4.6 ml of DMSO
was stirred at 80 C for 18 h. 0.77 ml of 2M aqueous sodium hydroxide solution was added, and the mixture was stirred at RT overnight. The mixture was diluted with water, adjusted to a pH of 3 - 4 with 10 percent strength aqueous citric acid solution and extracted three times with ethyl acetate, the extracts were concentrated and the residue was purified by preparative HPLC. This gave 7 mg of a solid. C281132FN03. MS (ESIpos) mass found: 449.24.

(400MHz, DMSO-d6): [ppm]= 0.98 (s, 314), 1.16 (s, 6H), 1.31 - 1.61 (m, 4H), 1.65 ¨ 1.76 (m, 114), 1.81 - 1.92 (m, 1H), 2.05 ¨ 2.38 (m, 511), 2.75 ¨ 2.84 (m, 2H), 3.85 (s, 2H), 6.24 -6.28 (m, 1H), 6.59 (d, 1H), 6.64 (dd, 111), 7.12 (d, 1H), 7.67 (dt, 1H), 8.43 (d, 114), 8.49 (t, 1H), 12.3 (s).
Example 9 4-([17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylloxylbutanoic acid N F

1.0 A mixture of 100 mg (0.29 mmol) of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 78 mg (0.43 mmol, 1.2 equiv.) of methyl 4-bromobutanoate, 198 mg (1.53 mmol, 5 .0 equiv.) of potassium carbonate and 4 mg of sodium iodide was stirred at 80 C for 18 h.
Another 78 mg of methyl 4-bromobutyrate were then added, and the mixture was stirred at overnight. 0.72 ml of 2M aqueous sodium hydroxide solution was added, and the mixture was stirred at 40 C for 3 h. The mixture was diluted with water, adjusted to a pH
of 4 with 10 percent strength aqueous citric acid solution and extracted three times with ethyl acetate, the extracts were concentrated and the residue was purified by preparative HPLC.
This gave 43 mg of a solid. C271130FN03. MS (ESIpos) mass found: 435.22. 1H-NMR (400MHz, DMSO-d6): 6 [ppm]= 0.99 (s, 3H), 1.31 - 1.61 (m, 4H), 1.71 (td, 111), 1.82 - 1.92 (m, 3H), 2.06 - 2.37 (m, 711), 2.77 - 2.84 (m, 2H), 3.89 (t, 2H), 6.25 (dd, 1H), 6.57 ¨ 6.81 (m, 1H), 6.62 - 6.68 (m, 1H), 7.12 (d, 114), 7.64 - 7.70 (m, 111), 8.43 (d, 1H), 8.48 (t, 1H), 12.1 (br. s, 1H).
Example 10 (RS)-2- ff17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yll oxy) propanoic acid N

CH3 404110, H0 )(LO

Analogously to Example 8, 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol were reacted with 117 mg (3.0 equiv.) of ethyl 2-chloropropanoate at 80 C
overnight. After addition of 2M aqueous sodium hydroxide solution, the mixture was stirred at 40 C for 2.5 h.

This gave, after preparative HPLC, 44 mg of the title compound. C26H28FN03. MS
(ESIpos) mass found: 421.21. 111-NMR (300MHz, DMSO-d6): [ppm]= 0.98 (s, 3H), 1.29 -1.62 (m, 8H), 1.70 (td, 1H), 1.79 ¨ 1.92 (m, 1H), 2.02 - 2.38 (m, 6H), 2.67 ¨ 2.86 (m, 2H), 4.70 (q, 1H), 6.26 (br. s., 1H), 6.49 - 6.63 (m, 2H), 7.12 (d, 1H), 7.67 (dt, 1H), 8.43 (d, 1H), 8.47 ¨
8.50 (m, 1H), 12.9 (br. s, 1H).
Example 11 (RS)-2-{ [17-(5-Fluoropyridin-3-yl)estra-1,3,5 (10),16-tetraen-3-yl] oxy} b utanoic acid N

HO O. 'III

Analogously to Example 8, 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol were reacted with 117 mg (3.0 equiv.) of methyl 2-chlorobutanoate at 80 C for 7 h and at room temperature for 72 h. After addition of 2M aqueous sodium hydroxide solution, the mixture was stirred at 40 C for 5.5 h. This gave, after preparative HPLC, 48 mg of the title compound. C27H30FN03. MS (ESIpos) mass found: 435.22. 1H-NMR (400MHz, DMSO-d6):
8 [ppm]= 0.95 (t, 3H), 0.99 (s, 3H), 1.31 - 1.61 (m, 4H), 1.63 - 1.92 (m, 4H), 2.09 - 2.37 (m, 5H), 2.69 - 2.89 (m, 2H), 4.48 - 4.55 (m, 1H), 6.22 - 6.28 (m, 1H), 6.52 ¨6.56 (m, 1H), 6.57 ¨
6.63 (m, 1H), 7.12 (d, 1H), 7.64 - 7.69 (m, 1H), 8.42 (d, 1H), 8.48 (t, 1H), 12.87 (br. s., 1H).
Example 12 (RS)-2- {117-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylloxy}-3-methoxy-propanoic acid F

CH

o HO SO
)r.0 Analogously to Example 8, 100 mg of 17-(5-fluoropyridin-3-ypestra-1,3,5(10),16-tetraen-3-ol were reacted with 169 mg (3.0 equiv.) of methyl 2-bromo-3-methoxypropanoate at 80 C for 4 h. After addition of 2M aqueous sodium hydroxide solution, the mixture was stirred at 40 C
for 5.5 h. This gave, after preparative HPLC, 57 mg of the title compound.
C27H30FN04. MS
(ESIpos) mass found: 451.22. 1H-NMR (400MHz, DMSO-d6): 6 [ppm]--- 0.99 (s, 3H), 1.32 -1.61 (m, 4H), 1.71 (td, 1H), 1.81 - 1.92 (m, 1H), 2.05 - 2.37 (m, 5H), 2.71 -2.87 (m, 2H), 3.28 (s, 3H), 3.66 - 3.78 (m, 2H), 4.79 ¨ 4.84 (m, 1H), 6.24 - 6.27 (m, 1H), 6.54 -6.59 (m, 1H), 6.62 (dt, 1H), 7.12 (d, 1H), 7.67 (dt, 1H), 8.42 (d, 111), 8.48 (t, 1H), 13.01 (hr. s., 111).
Example 13 (RS)-2- { [17-(3-Pyridyl)estra-1,3,5(10),16-tetraen-3-yl] oxy} p ropanoic acid N

HO (L

Analogously to Example 8, 100 mg of 17-(3-pyridyl)estra-1,3,5(10),16-tetraen-3-ol were reacted with 123 mg (3.0 equiv.) of ethyl 2-chloropropanoate at 80 C
overnight. After addition of 2M aqueous sodium hydroxide solution, the mixture was stirred at 40 C for 4 h.
This gave, after preparative HPLC, 19 mg of the title compound. C26H29NO3. MS
(ESIpos) mass found: 403.21. 11-1-NMR (400MHz, DMSO-d6): 6 [ppm]=-- 0.98 (s, 3H), 1.31 -1.61 (m, 7H, contains doublet at 1.43 ppm), 1.71 (td, 1H), 1.81 - 1.92 (m, 1H), 2.02 -2.15 (m, 2H), 2.15 - 2.38 (m, 3H), 2.69 - 2.89 (m, 211), 4.70 (qd, 1H), 6.11 (dd, 111), 6.51 - 6.55 (m, 1H), 6.59 (dt, 1H), 7.12 (d, 1H), 7.29 - 7.34 (m, 1H), 7.76 (dt, 1H), 8.41 (dd, 1H), 8.58 (d, 1H), 12.9 (br. s., 1H).
Example 14 (RS)-2-([17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylioxy}propanamide N F

1 _ CH
H2N ilia \ 3 A mixture of 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 92 mg (3.0 equiv.) of 2-chloropropanamide, 198 mg (5 equiv.) of potassium carbonate and 4 mg of sodium iodide in 3 ml DMSO was stirred at 80 C for 18 h, then at 100 C for 4 hand at 120 C
for 18 h. The mixture was diluted with water and extracted three times with ethyl acetate, and the combined organic phases were dried over sodium sulphate and concentrated.
Purification of the residue by preparative HPLC gave 18 mg of the title compound.
Purification method preparative HPLC:
System: Waters Autopurification system: Pump 254, Sample Manager 2767, CFO, DAD 2996, ELSD 2424, SQD 3100 Column: XBrigde C18 51.1m 100x30 mm Solvent: A = water + 0.1% vol. HCOOH (99%) B = acetonitrile Gradient: 0-8 min 50-90% B
Flow rate: 50 ml/min Temperature: RT
Detection: DAD scan range 210-400 nm MS ESI+, ESI-, scan range 160-1000 m/z ELSD
Fractions Rt in mm purity in % amount in peak assignment mg Example 14 4.4 ¨ 4.7 98.6 18 8 -1.41min Work-up: The fractions were evaporated, tBuOH was added, the fractions were frozen at -65 C and then freeze-dried.
C26H29FN202. MS (ESIpos) mass found: 420.22. 11-1-NMR (400MHz, DMSO-d6): 8 [ppm]=
0.99 (s, 3H), 1.31 - 1.61 (m, 7H, contains doublet at 1.37 ppm), 1.71 (td, 1H), 1.87 (dt, 1H), 2.06 - 2.38 (m, 511),.2.70 - 2.89 (m, 2H), 4.51 (q, 111), 6.24 - 6.27 (m, 1H), 6.58 (t, 1H), 6.64 (dt, 1H), 7.10 ¨ 7.18 (m, 211), 7.36 (d, 111), 7.63 - 7.71 (m, 1H), 8.43 (d, 1H), 8.46 - 8.51 (m, 1H).
Example 15 34[17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylloxylpropan-1-61 N---- F
\ /

0.
HOC) ..
A mixture of 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 119 mg (3.0 equiv.) of 3-bromopropan-1-ol, 198 mg (5 equiv.) of potassium carbonate and 4 mg of sodium iodide in 3 ml of DMSO was stirred at 80 C for 18 h, then at 100 C for 4 h.
The mixture was diluted with water and extract three times with ethyl acetate, and the combined organic phases were dried over sodium sulphate and concentrated. Purification of the residue by preparative HPLC gave 42 mg of the title compound. C26H30FN02. MS (ESIpos) mass found:
407.23. 11-1-NMR (300MHz, DMSO-d6): 8 [ppm]= 0.98 (s, 311), 1.29 - 1.62 (m, 4H), 1.64 -1.91 (m, 411), 2.03 - 2.39 (m, 5H), 2.71 - 2.90 (m, 211), 3.44 - 3.55 (m, 2H), 3.93 (t, 2H), 4.48 (t, 1H), 6.23 ¨
6.28 (m, 111), 6.59 (d, 1H), 6.64 (dd, 111), 7.11 (d, 111), 7.67 (dt, 1H), 8.43 (d, 1H), 8.46 ¨
8.51 (m, Hi).
Example 16 34[17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ylloxyl-2,2-dimethylpropan-1-ol = N F

HOC) .0 A mixture of 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 143 mg (3.0 equiv.) of 3-bromo-2,2-dimethylpropan-1-ol, 198 mg (5 equiv.) of potassium carbonate and 4 mg of sodium iodide in 3 ml of DMSO was stirred at 80 C for 18 h, then at 100 C for 4 h, at 120 C for 18 h and at 150 C for 5 h. The mixture was diluted with water and extracted three times with ethyl acetate, and the combined organic phases were dried over sodium sulphate and concentrated. Purification of the residue by preparative HPLC gave 42 mg of the title compound. C28H34FN02. MS (ESIpos) mass found: 435.26. 1H-NMR (400MHz, DMSO-d6):
8 [ppm]-= 0.87 (s, 6H), 0.99 (s, 3H), 1.31 - 1.61 (m, 4H), 1.71 (td, 1H), 1.83 - 1.91 (m, 1H), 2.06 - 2.37 (m, 5H), 2.73 - 2.87 (m, 2H), 3.23 (d, 2H), 3.60 (s, 2H), 4.52 (t, 1H), 6.26 (dd, 1H), 6.59 (d, 1H), 6.64 (dd, 1H), 7.11 (d, 1H), 7.64 - 7.70 (m, 1H), 8.43 (d, 1H), 8.48 (t, 111).
Example 17 (RS)-3-{(17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propan-1,2-diol N--- F

HOr0 OH
A mixture of 150 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 143 mg (3.0 equiv.) of 3-bromo-2,2-dimethylpropan-1-ol, 296 mg (5 equiv.) of potassium carbonate and 7 mg of potassium iodide in 3 nil of DMSO was stirred at 120 C for 18 h. The mixture was diluted with water and extracted three times with ethyl acetate, and the combined organic phases were dried over sodium sulphate and concentrated. Purification of the residue by preparative HPLC gave 26 mg of the title compound. C26H30FN03. MS (ESIpos) mass found:
435.26. 1H-NMR (300MHz, DMSO-d6): [ppm]= 0.99 (s, 3H), 1.30 - 1.62 (m, 4H), 1.71 (td, 1H), 1.81 ¨ 1.93 (d, 1H), 2.02 - 2.39 (m, 5H), 2.74 - 2.86 (m, 2H), 3.39 (t, 211), 3.67 - 3.80 (m, 2H), 3.90 (dd, 1H), 4.57 (t, 111), 4.83 (d, 1H), 6.26 (br. s., 1H), 6.55 -6.71 (m, 211), 7.12 (d, 111), 7.67 (dt, 1H), 8.40 - 8.52 (m, 2H).
Example 18 (RS)- ([17-(5-Fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl] oxy}
(phenyl)acetic acid N F

HO

A mixture of 100 mg of 17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-ol, 170 mg of ethyl chloro(phenyl)acetate (3 equiv.), 197 mg (5 equiv.) of potassium carbonate and 4 mg of sodium iodide in 3 ml of DMSO was stirred at 80 C for 3 days. 0.7 ml of 2M
aqueous sodium hydroxide solution was then added, and the mixture was stirred at 40 C for 5.5 h. The reaction mixture was diluted with water, acidified to pH = 4 with 10 percent strength citric acid solution and extracted three times with ethyl acetate. The combined organic phases were dried and concentrated. Purification by preparative HPLC gave 19 mg of the title compound.
Purification method preparative HPLC:
System: Agilent: Prep 1200, 2xPrep Pump, DLA, MWD, Prep FC, Column: XBrigde C18 5p.m 100x30 mm Solvent: A = water + 0.1% TFA
B = acetonitrile Gradient: 0-17.5 min 65-100% B, 17.5-20 min 100% B
Flow rate: 38 ml/min Temperature: RT
Detection: UV 254 nm Fractions:
Example 18 6.1 ¨ 6.7 min 96% 19 mg peak 12.2 min Work-up: The fractions were evaporated and then freeze-dried.
C311-130FN03.MS (ESIpos) mass found: 483.22. 1H-NMR (300MHz, DMSO-d6): 8 [ppm]=
0.98 (s, 3H), 1.30 - 1.63 (m, 4H), 1.70 (td, 111), 1.81 ¨ 1.91 (m, 1H), 2.04 -2.38 (m, 5H), 2.74 ¨ 2.84 (m, 211), 5.71 (s, 111), 6.23 ¨ 6.27 (m, 1H), 6.63 - 6.73 (m, 2H), 7.14 (d, 1H), 7.32 -* 7.42 (m, 3H), 7.48 - 7.54 (m, 2H), 7.64 - 7.70 (m, 1H), 8.42 (d, 1H), 8.48 (s, 1H), 13.1 (br. s., 1H).
Example 19 N-Ethyl-2-({17-[5-(trifluoromethyl)pyridin-3-yl] estra-1,3,5(10),16-tetraen-3-ylloxy)acetamide 00-1.
FI3CNC0 04o 66 mg (3 equiv.) of 1,1'-carbonyldiimidazole and 7 mg of imidazole hydrochloride were added to 62 mg (0.14 mmol) of ({17-[5-(trifluoromethyl)pyridin-3-yl]estra-1,3,5(10),16-tetraen-3-ylloxy)acetic acid in 3 ml of 2-methyltetrahydrofuran, and the mixture was stirred at room temperature overnight. 0.5 ml of DMF was added, and the mixture was heated at 50 C
for 6 h. 0.34 ml of a 2M ethylamine solution in THF and 57 microlitre of triethylamin were added, and the mixture was stirred at room temperature for 3 days. The reaction mixture was diluted with water and extracted three times with ethyl acetate, the combined organic phases were concentrated and the residue was purified by preparative HPLC. This gave 41 mg of the title compound. C28H31F3N202. MS (ESIpos) mass found: 484.23. 11-1-NMR
(300MHz, DMSO-d6): 8 [ppm]= 0.93 - 1.07 (m, 6H), 1.33 - 1.66 (m, 411), 1.73 (td, 114), 1.83 - 1.92 (m, 1H), 2.05 - 2.41 (m, 5H), 2.76 - 2.86 (m, 2H), 3.06 - 3.16 (m, 2H), 4.34 (s, 2H), 6.32 ¨ 6.38 (m, 1H), 6.62 - 6.72 (m, 2H), 7.15 (d, 111), 7.97 - 8.06 (m, 211), 8.81 ¨ 8.85 (m, 1H), 8.88 -8.92 (m, 111).
Pharmacological examination of the compounds according to the invention in vitro Example 20 (AKR1C3-inhibitory activity) The AKR1C3-inhibitory activity of the substances of the present invention was measured in the AKR1C3 assay described in the paragraphs below.

Essentially, the enzyme activity is measured by quantification of the Coumberol from Coumberone (Halim, M., Yee, D.J., and Sames, D., J. AM. CHEM. SOC. 130, 14123-(2008) and Yee, D.J., Balsanek, V., Bauman, D.R., Penning, T.M., and Sames, D., Proc. Natl.
Acad. Sci. USA 103, 13304 ¨ 13309 (2006)). In this test, the increase of the highly fluorescent Coumberol by NADPH- (nicotinamide adenine dinucleotide phosphate)-dependent reduction of the non-fluorescent Coumberone by AKR1C3 can be determined.
The enzyme used was recombinant human AKR1C3 (Aldo-keto reductase family 1 member C3) (GenBank Accession No. NM 003739). This was expressed in E. coli as GST
(glutathione S transferase) fusion protein and purified by glutathione Sepharose affinity chromatography. The GST was removed by digestion with thrombin and subsequent size exclusion chromatography (Dufort, I., Rheault, P., Huang, XF., Soucy, P., and Luu-The, V., Endocrinology 140, 568-574 (1999)).
For the assay, 50 n1 of a 100-fold concentrated solution of the test substance in DMSO were pipetted into a black low-volume 384-well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2.0 ul of a solution of AKR1C3 in assay buffer [50 mM potassium phosphate buffer pH 7, 1 mM DTT, 0.0022% (w/v) Pluronic F-127, 0.01% BSA (w/v) and protease inhibitor cocktail (Complete, EDTA-free Protease Inhibitor Cocktail from Roche)] were added and the mixture was incubated for 15 mM to allow pre-binding of the substances to the enzyme prior to the enzyme reaction. The enzyme reaction was then started by addition of 3 ul of a solution of NADPH (16.7 uM
final concentration in 5 ul of assay volume is 10 M) and Coumberone (0.5 uM final concentration in 5 ul of assay volume is 0.3 uM) in assay buffer, and the resulting mixture was incubated at 22 C for the reaction time of 90 mM.
The concentration of the AKR1C3 was adapted to the respective activity of the enzyme preparation and adjusted such that the assay was carried out in the linear range. Typical concentrations were in the region of 1 nM. The reaction was stopped by addition of 5 ul of a stop solution consisting of the inhibitor EM-1404 [F. Labrie et al. US Patent 6,541,463, 2003]
(2 M -3 final concentration in 5 1 of assay volume is 1 uM). The fluorescence of the Coumberole was then measured at 520 nm (excitation at 380 nm) using a suitable measuring instrument (Pherastar from BMG Labtechnologies). The intensity of the fluorescence was used as a measure of the amount of Coumberole formed and thus of the enzyme activity of AKR1C3. The data were normalized (enzyme reaction without inhibitor = 0 %
inhibition; all other assay components, but no enzyme = 100% inhibition). Usually, the test substances were tested on the same microtiter plate at 11 different concentrations in the range from 20 uM to 96.8 pM (20 uM, 5.9 uM, 1.7 uM, 0.5 M, 0.15 uM, 44 nM, 12.9 nM, 3.8 nM, 1.1 nM, 0.3 nM and 96.8 pM, the dilution series were prepared prior to the assay on the level of the 100-fold concentrated solution by serial 1:3 dilutions with 100% DMSO) in double for each concentration, and the IC50 values were calculated using a 4-parameter fit.
As described, the pharmacological substances claimed were examined for their inhibitory activity on the AKR1C3 enzyme (see Table 1). The compounds claimed show a strong inhibition of AKR1C3 in vitro (IC50 values < 200 nM) and in most cases even IC50 values <
50 nM.
Table 1: Inhibition of AKR1C3 of the compounds according to the invention (for some of the compounds, the values of two experimental determinations are stated) Example AKR1C3 enzyme Example AKR1C3 enzyme Example compound inhibition IC50 compound inhibition IC50 compound enzyme [nmo1/1]) [nmo1/1]) inhibition ICso [nmo1/1]) 1 7.6 6 9.2 13 2.8 1 8.0 7 1.4 14 20 3 8.6 10 2.7 18 10 4 136 11 2.5 19 53 5 89 12 4.0 Example 21 (test for AKR1C3 inhibition in a cell-based system) The inhibition of AKR1C3 by the substances described in the present invention was measured in a cell-based assay using coumberol as substrate for the AKR1C3 (Halim, M., Yee, D.J., and Sames, D., J. AM. CHEM. SOC. 130, 14123-14128 (2008) and Yee, D.J., Balsanek, V., Bauman, D.R., Penning, T.M., and Sames, D., Proc. Natl. Acad. Sci. USA 103, 13304 ¨
13309 (2006)) (cf. Example 20).
The cell system used were HEK293 cells (ATCC, USA) (cell culture medium: DMEM, 1.5 g glucose, 10% FCS, PSG). The cells were transfected in an AKR1C3 expression plasmid (pCMV6-AC-AKR1C3, GenBank Accession No. NM_003739.4) overnight (X-tremeGENE
HP, Roche). The next morning, the cells were sown in black 96we11 cell culture plates at a cell density of 40000 cells/well (Greiner Bio-One, Frickenhausen, Germany). After 7 h, the cells were incubated with the test substances (dissolved in 100x concentration in DMSO, final concentration between 10-11M and 10-5M) and coumberol (dissolved in cell culture medium, final concentration 5x10-6M) overnight. The next morning, the fluorescence of coumberol was measured at 535 nm (excitation at 355 nm) using a suitable measuring instrument (Mithras, from Berthold). The fluorescence intensity was used as a measure of the amount of coumberol formed and thus of the enzyme activity of AKR1C3. The data were normalized (transfected cells without inhibitor, only DMSO = 0% inhibition; transfected cells, 10 tM
inhibitor EM-1404 [F. Labrie et al. US Patent 6,541,463, 2003] = 100% inhibition), and the IC50 values were calculated using a 4-parameter fit.
In the cell-based assay described above, the pharmacological substances claimed were examined for their inhibitory activity on the AKR1C3 enzyme (see Table 2). The compounds showed a strong inhibition of cellular AKR1C3 in vitro (IC50 values < 300 nM) and in most cases even IC50 values < 100 nM.
Table 2: Inhibition of AKR1C3 of the compounds according to the invention (for some of the compounds, the values of independent experimental determinations are stated) Example compound Cellular AKR1C3 inhibition IC50 [nmo1/1]) Example 22 (AKR1C3 binding) Binding of Example compound 1 to the AKR1C3 enzyme was measured by isothermal titration calorimetry. The degree of affinity of a given compound can be stated using the dissociation constant (KD value). Human AKR1C3 enzyme was prepared as described in Example 20. For ITC experiments, human AKR1C3 protein was transferred by gel permeation into a PBS buffer consisting of 137 mM NaC1, 2.7 mM KC1, 10 mM
Na2HPO4, 2mM KH2PO4 and 1 mM tris(2-carboxyethyl)phosphine hydrochloride. The compounds were dissolved in 100% DMSO at a concentration of 10 mM and then diluted with PBS
buffer.
Isothermal titration calorimetry experiments (ITC experiments) were carried out using an ITC200 titration calorimeter (GE Healthcare, Northampton, MA, USA) at a temperature of 25 C. The change in enthalpy resulting from the injection of the protein was determined by integration of the calorimetric signal. The data were analyzed with Origin 7.0 (GE Healthcare, Northampton, MA, USA). Heat of dilution values were determined using the last injection of the titration in question and subtracted prior to curve fitting. The protein concentration in the injection syringe was in the range from 80 to 120 1.1.M and the concentration of the compound to be measured was in the range from 5 to 20 M.
A KD value of 230 nanomolar and an exothermal binding enthalpy of -1500 kcal/mol were determined for Example compound 1.

Claims (13)

claims
1. Compounds of the formula (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyrimidin-4-yl, pyrazin-2-yl, pyridazin-4-yl, pyridazin-3-yl, optionally mono- or disubstituted by fluorine, chlorine, nitrile, hydroxyl, carboxyl, trifluoromethyl, pentafluoroethyl, methoxy, ethoxy, trifluoromethoxy, -OCH2CF3, -SO2CH3, -SO2CH2CH3, -(C=O)CH3, C1-C4-Alkyl, -CH2OH, -C(CH3)2OH, -CONH2, -(C=O)NHCH3, -(C=O)NHCH2CH3, -(C=O)N(CH3)2, -SO2NH2, -SO2NHCH3, -SO2N(CH3)2, R1 represents -O-CR a R b -Y where R a and R b independently of one another represent represent hydrogen, methyl, ethyl, propyl, isopropyl, phenyl, 4-fluorophenyl, 3-fluorophenyl, 2-fluorophenyl, CH3-O-CH2-, -CH2CF3 or R a and R b together represent -(CH2)n- where n = 2, 3, 4 or 5 or R a and R b together represent -CH2-O-CH2-, -CH2-NH-CH2-, -CH2-N(CH3)-CH2-, -CH2CH2-O-CH2CH2-, -CH2CH2-NH-CH2CH2-, -CH2CH2-N(CH3)-CH2CH2--O-CR c R d-CR e R f-y where R e, R d, R e R f represent hydrogen or R e, R f represent hydrogen and R c, R d independently of one another represent methyl, ethyl or together represent -(CH2)n- where n = 2, 3, 4, or together represent -CH2-O-CH2- or -CH2CH2-O-CH2CH2- or R c, R d represent hydrogen and R e, R f independently of one another . represent methyl, ethyl, CF3CH2- or together represent -(CH2)n- where n = 2, 3, 4, 5, -CH2CH2-O-CH2CH2-, -CH2CH2-NH-CH2CH2-, -CH2CH2-N(CH3)-CH2CH2-, -CH2-O-CH2-, -CH2-NH-CH2-, -CH2-N(CH3)-CH2- or R d, R e, R f represent hydrogen and R c represents methyl, ethyl, trifluoromethyl or R c, R d, R f represent hydrogen and R e represents methyl, ethyl, trifluoromethyl, hydroxyl, methoxy, trifluoromethoxy or R d, R f represent hydrogen and R c, R e independently of one another represent methyl, ethyl, trifluoromethyl, -O-CH2CH2CH2-Y, -O-CH2C(CH3)2CH2-Y, -O-CH2CH2C(CH3)2-Y, -O-CH2CH2CH(CH3)-Y, -O-CH2-CH(OH)-CH2-Y
-OCH2CH2CH2CH2-Y, -CH2-Y, -CR g R h-CR i R j-Y where R g, R h, R i, R j represent hydrogen or R g, R h, R i represent hydrogen and R j represents methyl, ethyl, trifluoromethyl or R i, R j represent hydrogen and R g, R h represent methyl or together represent -CH2-, -CH2CH2-, -CH2CH2CH2- or R g represents methyl and R h, R i, R j represent hydrogen, -CH2CH2CH2-Y, -CH2CH2C(CH3)2-Y or -CH2CH2CH2CH2-Y and Y represents -CO2H, -OH, -(C=O)NH2, -(C=O)NHC1-4-alkyl, -S(=O)CH3 and their salts, solvates and solvates of the salts.
2. Compounds o.f the formula (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyrimidin-4-yl, pyrazin-2-yl, pyridazin-4-yl, pyridazin-3-yl, optionally monosubstituted by fluorine, chlorine, nitrile, hydroxyl, carboxyl, trifluoromethyl, methoxy, ethoxy, trifluoromethoxy, -SO2CH3, -(C=O)CH3, C1-C4-alkyl, R1 represents -O-CR a R b -Y where R a and R b independently of one another represent represent hydrogen, methyl, ethyl, propyl, isopropyl, phenyl, CH3-O-CH2-, CF3CH2-, -O-CR c R d-CR e R f-Y where R c, R d, R e R f represent hydrogen or R c, R d represent hydrogen and R e, R f independently of one another represent methyl, ethyl, CF3CH2- or R d, R e, R f represent hydrogen and R c represents methyl, ethyl or R c, R d, R f represent hydrogen and Re represents methyl, ethyl, -O-CH2CH2CH2-Y, -O-CH2C(CH3)2CH2-Y, -O-CH2CH2C(CH3)2-Y, -O-CH2CH2CH(CH3)-Y, -O-CH2-CH(OH)-CH2-Y or -CR g R h-CR i R j-Y where R g, R h, R i, R j represent hydrogen or R g, R h, R i represent hydrogen and R j represents methyl, ethyl or R i, R j represent hydrogen and R g, R h represents methyl or R g represents methyl and R h, R i, R j represent hydrogen and Y represents -CO2H, -OH, -(C=O)NH2, -(C=O)NHC1-4-alkyl and their salts, solvates and solvates of the salts.
3. Compounds of the formula (I) in which A represents pyridin-3-yl, pyrimidin-5-yl, pyridazin-4-yl, optionally monosubstituted by fluorine, carboxyl, trifluoromethyl, methyl R1 represents -O-CR a R b -Y where R a and R b represent hydrogen or R a represents hydrogen and R b represents methyl, ethyl, phenyl or CH3-O-CH2--O-CR c R d-CR e R f-Y where R c, R d represent hydrogen and R e, R f represent methyl -O-CH2CH2CH2-Y, -O-CH2C(CH3)2CH2-Y, -O-CH2-CH(OH)-CH2-Y or -CH2-CH2-Y and Y represents -CO2H, -OH, -(C=O)NH2, -(C=O)NHC1-4-alkyl and their salts, solvates and solvates of the salts.
4. Compounds of the formula (I) in which A represents 5-fluoropyridin-3-yl, 5-(trifluoromethyl)pyridin-3-yl, 5-carboxy-pyridin-3-yl, 6-methylpyridin-3-yl, pyrimidin-5-yl, 6-methylpyridazin-4-yl and R1 represents -O-CR a R b -CO2H, -O-CR a R b-(C=O)NH2, -O-CR a R b-(C=O)NHCH2CH3 where R a and R b represent hydrogen or R a represents hydrogen and R b represents methyl, ethyl, phenyl, CH3OCH2-, -O-CR c R d-CR e R f-CO2H where R c, R d represent hydrogen and R e, R f represent methyl, -O-CH2CH2CH2-OH, -O-CH2CH2CH2-CO2H, -O-CH2C(CH3)2CH2-OH or -O-CH2-CH(OH)-CH2-OH

and their salts, solvates and solvates of the salts.
5. Compounds of the formula (I), namely 3-[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]propanoic acid ({17-[5-(trifluoromethyl)pyridin-3-yl]estra-1,3,5(10), 16-tetraen-3-yl}oxy)acetic acid {[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxylacetic acid {[17-(6-methylpyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}acetic acid {[17-(pyrimidin-5-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}acetic acid 5-[3-(carboxymethoxy)estra-1,3,5(10),16-tetraen-17-yl]nicotinic acid {[17-(6-methylpyridazin-4-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}acetic acid 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}-2,2-dimethylpropanoic acid 4-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}butanoic acid 2-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propanoic acid 2-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}butanoic acid 2-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}-3-methoxypropanoic acid 2-{[17-(3-pyridyl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propanoic acid 2-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propanamide 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propan-1-ol 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}-2,2-dimethylpropan-1-ol 3-{[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxy}propan-1,2-diol {[17-(5-fluoropyridin-3-yl)estra-1,3,5(10),16-tetraen-3-yl]oxyl(phenyl)acetic acid N-ethyl-2-({17-[5-(trifluoromethyl)pyridin-3-yl]estra-1,3,5(10),16-tetraen-3-yl}oxy)acetamide and their salts, solvates and solvates of the salts.
6. Compound of the formula (I) as defined in any of Claims 1 to 5 for the treatment and/or prophylaxis of diseases.
7. Compound of the formula (I) as defined in any of Claims 1 to 5 for use in a method for the treatment and/or prophylaxis of diseases of endometriosis, of uterine leiomyomas, of uterine bleeding disorders, of dysmenorrhoea, of prostate carcinoma, of prostate hyperplasia, of acne, of seborrhoea, of hair loss, of premature sexual maturity, of polycystic ovary syndrome, of breast cancer, of lung cancer, of endometrial carcinoma, of renal cell carcinoma, of bladder carcinoma, of non-Hodgkin lymphomas, of chronic obstructive pulmonary disease (COPD), of adiposity or of inflammatory pain.
8. Medicament comprising a compound of the formula (I) as defined in any of Claims 1 to 5 in combination with one or more further active compound(s).
9. Medicament comprising a compound of the formula (I) as defined in any of Claims 1 to 5 in combination with an inert non-toxic pharmaceutically suitable auxiliary.
10. Medicament according to Claim 8 or 9 for the treatment and/or prophylaxis of endometriosis, of uterine leiomyomas, of uterine bleeding disorders, of dysmenorrhoea, of prostate carcinoma, of prostate hyperplasia, of acne, of seborrhoea, of hair loss, of premature sexual maturity, of polycystic ovary syndrome, of breast cancer, of lung cancer, of endometrial carcinoma, of renal cell carcinoma, of bladder carcinoma, of non-Hodgkin lymphomas, of chronic obstructive pulmonary disease (COPD), of adiposity or of inflammatory pain.
11. Method for the treatment of endometriosis in humans and animals by administration of an antiproliferatively effective amount of at least one compound of the formula (I) as defined in any of Claims 1 to 5 or of a medicament as defined in any of Claims 8 to 10.
12. Method for the treatment of endometriosis in humans and animals by administration of an anti-inflammatorily effective amount of at least one compound of the formula (I) as defined in any of Claims 1 to 5 or of a medicament as defined in any of Claims 8 to 10.
13. Method for the treatment of endometriosis in humans and animals by administration of an antinociceptively effective amount of at least one compound of the formula (I) as defined in any of Claims 1 to 5 or of a medicament as defined in any of Claims 8 to 10.
CA2878485A 2012-07-10 2013-07-05 3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments Abandoned CA2878485A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE102012211970.1 2012-07-10
DE102012211970 2012-07-10
PCT/EP2013/064257 WO2014009274A1 (en) 2012-07-10 2013-07-05 3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments

Publications (1)

Publication Number Publication Date
CA2878485A1 true CA2878485A1 (en) 2014-01-16

Family

ID=48748225

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2878485A Abandoned CA2878485A1 (en) 2012-07-10 2013-07-05 3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments

Country Status (28)

Country Link
US (1) US9512169B2 (en)
EP (1) EP2872524B1 (en)
JP (1) JP6196302B2 (en)
KR (1) KR20150036319A (en)
CN (1) CN104428309B (en)
AP (1) AP2014008179A0 (en)
AR (1) AR092025A1 (en)
AU (1) AU2013289344A1 (en)
BR (1) BR112015000443A2 (en)
CA (1) CA2878485A1 (en)
CL (1) CL2015000039A1 (en)
CO (1) CO7160008A2 (en)
CR (1) CR20150007A (en)
DO (1) DOP2015000005A (en)
EA (1) EA201500112A1 (en)
EC (1) ECSP15000868A (en)
ES (1) ES2603258T3 (en)
HK (1) HK1205743A1 (en)
IL (1) IL236581A0 (en)
MA (1) MA37742B1 (en)
MX (1) MX2015000347A (en)
PE (1) PE20150353A1 (en)
PH (1) PH12015500062A1 (en)
TN (1) TN2015000007A1 (en)
TW (1) TW201402596A (en)
UY (1) UY34899A (en)
WO (1) WO2014009274A1 (en)
ZA (1) ZA201500920B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210283151A1 (en) * 2018-07-09 2021-09-16 Ascentawits Pharmaceuticals, Ltd. Use of compound in drug for preventing, treating, or alleviating pain

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105008382B (en) * 2013-02-21 2018-01-30 拜耳医药股份公司 For suppressing the female steroid 1,3,5 (10) of 17 11-beta-hydroxysteroid dehydrogenase types (AKR1 C3), the carboxylic acid amides of 16 tetraene 3
JP6407415B2 (en) * 2014-09-11 2018-10-17 バイエル・ファルマ・アクティエンゲゼルシャフト 3-nitrogen or sulfur substituted estra-1,3,5 (10), 16-tetraene AKR1C3 inhibitor
US10167293B2 (en) 2016-05-26 2019-01-01 Bayer Pharma Aktiengesellschaft [8-(phenylsulfonyl)-3,8-diazabicyclo[3.2.1]oct-3-yl](1H-1,2,3-triazol-4-yl)methanones
CZ307437B6 (en) 2016-06-07 2018-08-22 Ăšstav organickĂ© chemie a biochemie AV ÄŚR, v.v.i. 15β-substituted estrone derivatives as selective inhibitors of 17β-hydroxysteoid dehydrogenases
EP3269373A1 (en) 2016-07-11 2018-01-17 Bayer Pharma Aktiengesellschaft Low-dosed oral dosage form of an akr1c3 inhibitor for treatment of diseases
TW201825478A (en) 2016-12-19 2018-07-16 德商拜耳製藥公司 [4-(phenylsulfonyl)piperazin-1-yl](1h-1,2,3-triazol-4-yl)methanones
EP3555067A1 (en) 2016-12-19 2019-10-23 Bayer Pharma Aktiengesellschaft [4-(phenylsulfonyl)piperazin-1-yl](1h-1,2,3-triazol-4-yl)methanones
WO2018114677A2 (en) 2016-12-19 2018-06-28 Bayer Pharma Aktiengesellschaft [(phenylsulfonyl)octahydro-epiminoisoindol-yl](1h-1,2,3-triazol-5-yl)methanones
EP3421483A1 (en) 2017-06-29 2019-01-02 Bayer Pharma Aktiengesellschaft Novel steroidal 17-beta heteroaryl compounds as inhibitors of akr1c3
CN110272465A (en) * 2019-07-15 2019-09-24 成都贝诺科成生物科技有限公司 Abiraterone derivative, preparation method and application

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5604213A (en) 1992-03-31 1997-02-18 British Technology Group Limited 17-substituted steroids useful in cancer treatment
NZ249911A (en) * 1992-03-31 1996-06-25 British Tech Group 17-(3-pyridyl) substituted steroids and use in pharmaceutical compositions
IL122740A (en) 1997-01-15 2003-09-17 Akzo Nobel Nv 16-hydroxy-11-(substituted phenyl)-estra-9,4-diene derivatives, their preparation and pharmaceutical compositions containing them
PT973790E (en) 1997-04-07 2003-10-31 Wyeth Corp ESTRA-5, 7-DIENES WITH ESTROGENIC ACTIVITY
US6541463B1 (en) * 1998-03-11 2003-04-01 Endorecherche, Inc. Inhibitors of type 5 and type 3 17β-hydroxysteroid dehydrogenase and methods for their use
AU5144999A (en) 1998-08-07 2000-02-28 Endorecherche Inc. Inhibition of type 3 3alpha-hydroxysteroid dehydrogenase
JPWO2001081364A1 (en) * 2000-04-24 2004-01-08 協和醗酵工業株式会社 Estra-1,3,5 (10) -triene derivative
US7498322B2 (en) 2004-03-12 2009-03-03 Entremed, Inc. Antiangiogenic agents
US7855225B2 (en) 2006-03-02 2010-12-21 Astellas Pharma Inc. 17βHSD type 5 inhibitor
MX2009005201A (en) 2006-11-30 2009-05-27 Solvay Pharm Gmbh Substituted estratrien derivatives as 17beta hsd inhibitors.
KR20090082502A (en) 2006-12-22 2009-07-30 에프. 호프만-라 로슈 아게 Spiro-piperidine derivatives
JP5251876B2 (en) 2007-07-24 2013-07-31 アステラス製薬株式会社 Benzimidazole derivatives
DE102011083725A1 (en) * 2011-09-29 2013-04-04 Bayer Pharma AG Estra-1,3,5 (10), 16-tetraene-3-carboxamide derivatives, process for their preparation, pharmaceutical preparations containing them and their use for the preparation of medicaments

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210283151A1 (en) * 2018-07-09 2021-09-16 Ascentawits Pharmaceuticals, Ltd. Use of compound in drug for preventing, treating, or alleviating pain

Also Published As

Publication number Publication date
ES2603258T3 (en) 2017-02-24
TN2015000007A1 (en) 2016-06-29
IL236581A0 (en) 2015-02-26
JP2015522050A (en) 2015-08-03
US9512169B2 (en) 2016-12-06
AR092025A1 (en) 2015-03-18
MA37742B1 (en) 2016-12-30
US20150210734A1 (en) 2015-07-30
AP2014008179A0 (en) 2014-12-31
WO2014009274A1 (en) 2014-01-16
ECSP15000868A (en) 2016-01-29
MA37742A1 (en) 2016-05-31
CO7160008A2 (en) 2015-01-15
EP2872524A1 (en) 2015-05-20
AU2013289344A1 (en) 2015-02-05
CN104428309A (en) 2015-03-18
KR20150036319A (en) 2015-04-07
EP2872524B1 (en) 2016-08-24
ZA201500920B (en) 2017-01-25
HK1205743A1 (en) 2015-12-24
UY34899A (en) 2014-02-28
DOP2015000005A (en) 2015-02-15
PE20150353A1 (en) 2015-03-28
EA201500112A1 (en) 2015-07-30
JP6196302B2 (en) 2017-09-13
CR20150007A (en) 2015-03-13
CL2015000039A1 (en) 2015-03-27
CN104428309B (en) 2016-09-14
BR112015000443A2 (en) 2017-06-27
TW201402596A (en) 2014-01-16
PH12015500062A1 (en) 2015-03-02
MX2015000347A (en) 2015-04-14

Similar Documents

Publication Publication Date Title
US9512169B2 (en) 3-substituted estra-1,3,5(10),16-tetraene derivatives, methods for the production thereof, pharmaceutical preparations containing same, and use thereof for the production of medicaments
CA2901632C (en) Estra-1,3,5(10),16-tetraene-3-carboxamides for inhibition of 17.beta.-hydroxysteroid dehydrogenase (akr1 c3)
CN103370322B (en) The Imidazopyrazines that the 2-being used as MPS-1 and TKK inhibitor in the treatment of hyperproliferative disorders replaces
JP6088542B2 (en) Substituted imidazopyrazines as Akt kinase inhibitors
WO2018114783A1 (en) Tetrazole containing compounds
TW201742868A (en) [8-(phenylsulfonyl)-3,8-diazabicyclo[3.2.1]oct-3-yl](1H-1,2,3-triazol-4-yl)methanones
AU2013283543B2 (en) N-[4-(Quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
WO2017025493A1 (en) Quinoline ezh2 inhibitors
WO2019002015A1 (en) Novel steroidal 17-beta heteroaryl compounds as inhibitors of akr1c3
JP6407415B2 (en) 3-nitrogen or sulfur substituted estra-1,3,5 (10), 16-tetraene AKR1C3 inhibitor

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20180215

FZDE Discontinued

Effective date: 20200831